1
|
Ulbrich M, Seward CH, Ivanov AI, Ward BM, Butler JS, Dziejman M. VopX, a novel Vibrio cholerae T3SS effector, modulates host actin dynamics. mBio 2025:e0301824. [PMID: 39878476 DOI: 10.1128/mbio.03018-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Pathogenic Vibrio cholerae strains cause cholera using different mechanisms. O1 and O139 serogroup strains use the toxin-co-regulated pilus (TCP) and cholera toxin (CT) for intestinal colonization and to promote secretory diarrhea, while non-O1/non-O139 serogroup strains are typically non-toxigenic and use alternate virulence factors to cause a clinically similar disease. An O39 serogroup, TCP/CT-negative V. cholerae strain, named AM-19226, uses a type III secretion system (T3SS) to translocate more than 10 effector proteins into the host cell cytosol. Effectors VopF and VopM directly interact with the host actin and contribute to colonization. Our previous studies using the Saccharomyces cerevisiae model system identified VopX as a third effector that alters cytoskeletal dynamics. Herein, we used complementary approaches to translate yeast findings to a mammalian system and determined the target and mechanism of VopX activity. VopX overexpression in HeLa cells caused dramatic cell rounding. Co-culture of strain AM-19226 with polarized Caco-2/BBE monolayers increased formation of stress fibers and focal adhesions, as well as Caco-2/BBE adherence to extracellular matrix in a VopX-dependent manner. Finally, we demonstrate in vitro that VopX can act as a guanine nucleotide exchange factor for RhoA, which functions upstream of a mitogen-activated protein kinase (MAPK) signaling pathway regulating cytoskeletal dynamics. Our results suggest that VopX activity initiates a signaling cascade resulting in enhanced cell-extracellular matrix adhesion, potentially preventing detachment of host cells, and facilitating sustained bacterial colonization during infection. VopX function is therefore part of a unique pathogenic strategy employed by T3SS-positive V. cholerae, which involves multiple cytoskeletal remodeling mechanisms to support a productive infection. IMPORTANCE Despite different infection strategies, enteric pathogens commonly employ a T3SS to colonize the human host and cause disease. Effector proteins are unique to each T3SS-encoding bacterial species and generally lack conserved amino acid sequences. However, T3SS effectors from diverse pathogens target and manipulate common host cell structures and signaling proteins, such as the actin cytoskeleton and MAPK pathway components. T3SS-encoding Vibrio cholerae strains and effectors have been relatively recently identified, and the mechanisms used to mediate colonization and secretory diarrhea are poorly understood. Two V. cholerae effectors that modify the host actin cytoskeleton were shown to be important for colonization. We therefore sought to determine the target(s) and mechanism of a third actin-reorganizing effector, VopX, based on results obtained from a yeast model system. We recapitulated actin-based phenotypes in multiple mammalian model systems, leading us to identify the molecular function of the V. cholerae VopX effector protein.
Collapse
Affiliation(s)
- Megan Ulbrich
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Christopher H Seward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Brian M Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - J Scott Butler
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Michelle Dziejman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
2
|
Pereiro P, Tur R, García M, Figueras A, Novoa B. Unravelling turbot ( Scophthalmus maximus) resistance to Aeromonas salmonicida: transcriptomic insights from two full-sibling families with divergent susceptibility. Front Immunol 2024; 15:1522666. [PMID: 39712009 PMCID: PMC11659141 DOI: 10.3389/fimmu.2024.1522666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Furunculosis, caused by the gram-negative bacterium Aeromonas salmonicida subsp. salmonicida, remains a significant threat to turbot (Scophthalmus maximus) aquaculture. Identifying genetic backgrounds with enhanced disease resistance is critical for improving aquaculture health management, reducing antibiotic dependency, and mitigating economic losses. Methods In this study, five full-sibling turbot families were challenged with A. salmonicida, which revealed one family with significantly greater resistance. Transcriptomic analyses (RNA-Seq) were performed on resistant and susceptible families, examining both naïve and 24-h postinfection (hpi) samples from head kidney and liver tissues. Results In the absence of infection, differentially expressed genes (DEGs) were identified predominantly in the liver. Following infection, a marked increase in DEGs was observed in the head kidney, with many genes linked to immune functions. Interestingly, the resistant family displayed a more controlled inflammatory response and upregulation of genes related to antigen presentation and T-cell activity in the head kidney at early infection stages, which may have contributed to its increased survival rate. In the liver, transcriptomic differences between the families were associated mainly with cytoskeletal organization, cell cycle regulation, and metabolic processes, including insulin signalling and lipid metabolism, regardless of infection status. Additionally, many DEGs overlapped with previously identified quantitative trait loci (QTLs) associated with resistance to A. salmonicida, providing further insights into the genetic basis of disease resistance. Discussion This study represents the first RNA-Seq analysis comparing resistant and susceptible turbot families and contributes valuable knowledge for the development of selective breeding programs targeting disease resistance in turbot and other aquaculture species susceptible to A. salmonicida.
Collapse
Affiliation(s)
- Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| | - Ricardo Tur
- Nueva Pescanova Biomarine Center, S.L., O Grove, Spain
| | - Miguel García
- Nueva Pescanova Biomarine Center, S.L., O Grove, Spain
| | - Antonio Figueras
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, Spain
| |
Collapse
|
3
|
Leal Y, Valenzuela-Muñoz V, Gallardo-Escárate C. Alternative splicing in Atlantic salmon head kidney and SHK-1 cell line during the Piscirickettsia salmonis infection: A comparative transcriptome survey. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109127. [PMID: 37813155 DOI: 10.1016/j.fsi.2023.109127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
Piscirickettsia salmonis, an intracellular bacterium in salmon aquaculture, is a big challenge because it is responsible for 54.2% of Atlantic salmon mortalities. In recent years, the high relevance of Alternative Splicing (AS) as a molecular mechanism associated with infectious conditions and host-pathogen interaction processes, especially in host immune activation, has been observed. Several studies have highlighted the role of AS in the host's immune response during viral, bacterial, and endoparasite infection. In the present study, we evaluated AS transcriptome profiles during P. salmonis infection in the two most used study models, SHK-1 cell line and salmon head kidney tissue. First, the SHK-1 cell line was exposed to P. salmonis infection at 0-, 7-, and 14-days post-infection (dpi). Following, total RNA was extracted for Illumina sequencing. On the other hand, RNA-Seq datasets of Atlantic salmon head kidney infected with the same P. salmonis strayingwase used. For both study models, the highest number of differentially alternative splicing (DAS) events was observed at 7 dpi, 16,830 DAS events derived from 9213 DAS genes in SHK-1 cells, and 13,820 DAS events from 7684 DAS genes in salmon HK. Alternative first exon (AF) was the most abundant AS type in the three infection times analyzed, representing 31% in SHK-1 cells and 228.6 in salmon HK; meanwhile, mutually exclusive exon (MX) was the least abundant. Notably, functional annotation of DAS genes in SHK-1 cells infected with P. salmonis showed a high presence of genes related to nucleotide metabolism. In contrast, the salmon head kidney exhibited many GO terms associated with immune response. Our findings reported the role of AS during P. salmonis infection in Atlantic salmon. These studies would contribute to a better understanding of the molecular bases that support the pathogen-host interaction, evidencing the contribution of AS regulating the transcriptional host response.
Collapse
Affiliation(s)
- Yeny Leal
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, P.O. Box 160-C, Concepción, 4030000, Chile.
| |
Collapse
|
4
|
Wang S, Hao J, Yang J, Zhang Q, Li A. The Attenuation Mechanism and Live Vaccine Potential of a Low-Virulence Edwardsiella ictaluri Strain Obtained by Rifampicin Passaging Culture. J Microbiol Biotechnol 2023; 33:167-179. [PMID: 36734130 PMCID: PMC9998210 DOI: 10.4014/jmb.2210.10013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 02/04/2023]
Abstract
The rifampicin-resistant strain E9-302 of Edwardsiella ictaluri strain 669 (WT) was generated by continuous passage on BHI agar plates containing increasing concentrations of rifampicin. E9-302 was attenuated significantly by 119 times to zebrafish Danio rerio compared to WT in terms of the 50% lethal dose (LD50). Zebrafish vaccinated with E9-302 via intraperitoneal (IP) injection at a dose of 1 × 103 CFU/fish had relative percentage survival (RPS) rates of 85.7% when challenged with wild-type E. ictaluri via IP 14 days post-vaccination (dpv). After 14 days of primary vaccination with E9-302 via immersion (IM) at a dose of 4 × 107 CFU/ml, a booster IM vaccination with E9-302 at a dose of 2 × 107 CFU/ml exhibited 65.2% RPS against challenge with wild-type E. ictaluri via IP 7 days later. These results indicated that the rifampicin-resistant attenuated strain E9-302 had potential as a live vaccine against E. ictaluri infection. A previously unreported amino acid site change at position 142 of the RNA polymerase (RNAP) β subunit encoded by the gene rpoB associated with rifampicin resistance was identified. Analysis of the whole-genome sequencing results revealed multiple missense mutations in the virulence-related genes esrB and sspH2 in E9-302 compared with WT, and a 189 bp mismatch in one gene, whose coding product was highly homologous to glycosyltransferase family 39 protein. This study preliminarily explored the molecular mechanism underlying the virulence attenuation of rifampicin-resistant strain E9-302 and provided a new target for the subsequent study of the pathogenic mechanism of E. ictaluri.
Collapse
Affiliation(s)
- Shuyi Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jingwen Hao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jicheng Yang
- Dalian Ocean University, Dalian 116023, P.R. China
| | - Qianqian Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P.R. China
| | - Aihua Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
5
|
Qin W, Xie Y, Ren Z, Xu C, Sun MA, Yin Z, Bao W. Integrative ATAC-seq and RNA-seq analyses of IPEC-J2 cells reveals porcine transcription and chromatin accessibility changes associated with Escherichia coli F18ac inhibited by Lactobacillus reuteri. Front Microbiol 2023; 14:1101111. [PMID: 36876070 PMCID: PMC9978113 DOI: 10.3389/fmicb.2023.1101111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Escherichia coli is the main cause of postweaning diarrhea in pigs, leading to economic loss. As a probiotic, Lactobacillus reuteri has been used to inhibit E. coli in clinical applications; however, its integrative interactions with hosts remain unclear, especially in pigs. Here, we found that L. reuteri effectively inhibited E. coli F18ac adhering to porcine IPEC-J2 cells, and explored the genome-wide transcription and chromatin accessibility landscapes of IPEC-J2 cells by RNA-seq and ATAC-seq. The results showed that some key signal transduction pathways, such as PI3K-AKT and MAPK signaling pathways, were enriched in the differentially expressed genes (DEGs) between E. coli F18ac treatment with and without L. reuteri groups. However, we found less overlap between RNA-seq and ATAC-seq datasets; we speculated that this might be caused by histones modification through ChIP-qPCR detection. Furthermore, we identified the regulation of the actin cytoskeleton pathway and a number of candidate genes (ARHGEF12, EGFR, and DIAPH3) that might be associated with the inhibition of E. coli F18ac adherence to IPEC-J2 cells by L. reuteri. In conclusion, we provide a valuable dataset that can be used to seek potential porcine molecular markers of E. coli F18ac pathogenesis and L. reuteri antibacterial activity, and to guide the antibacterial application of L. reuteri.
Collapse
Affiliation(s)
- Weiyun Qin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yunxiao Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhanshi Ren
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chao Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming-An Sun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
6
|
Kudryashova E, Ankita, Ulrichs H, Shekhar S, Kudryashov DS. Pointed-end processive elongation of actin filaments by Vibrio effectors VopF and VopL. SCIENCE ADVANCES 2022; 8:eadc9239. [PMID: 36399577 PMCID: PMC9674292 DOI: 10.1126/sciadv.adc9239] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/03/2022] [Indexed: 07/20/2023]
Abstract
According to the cellular actin dynamics paradigm, filaments grow at their barbed ends and depolymerize predominantly from their pointed ends to form polar structures and do productive work. We show that actin can elongate at the pointed end when assisted by Vibrio VopF/L toxins, which act as processive polymerases. In cells, processively moving VopF/L speckles are inhibited by factors blocking the pointed but not barbed ends. Multispectral single-molecule imaging confirmed that VopF molecules associate with the pointed end, actively promoting its elongation even in the presence of profilin. Consequently, VopF/L can break the actin cytoskeleton's polarity by compromising actin-based cellular processes. Therefore, actin filament design allows processive growth at both ends, which suggests unforeseen possibilities for cellular actin organization, particularly in specialized cells and compartments.
Collapse
Affiliation(s)
- Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Ankita
- Department of Physics, Emory University, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Heidi Ulrichs
- Department of Physics, Emory University, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Shashank Shekhar
- Department of Physics, Emory University, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Lactobacilli, a Weapon to Counteract Pathogens through the Inhibition of Their Virulence Factors. J Bacteriol 2022; 204:e0027222. [PMID: 36286515 PMCID: PMC9664955 DOI: 10.1128/jb.00272-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To date, several studies have reported an alarming increase in pathogen resistance to current antibiotic therapies and treatments. Therefore, the search for effective alternatives to counter their spread and the onset of infections is becoming increasingly important.
Collapse
|
8
|
Hussein H, Kishen A. Application of Proteomics in Apical Periodontitis. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.814603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Apical periodontitis is an inflammatory reaction of the periradicular tissues as a consequence of multispecies microbial communities organized as biofilms within the root canal system. Periradicular tissue changes at the molecular level initiate and orchestrate the inflammatory process and precede the presentation of clinical symptoms. Inflammatory mediators have been studied at either the proteomic, metabolomic, or transcriptomic levels. Analysis at the protein level is the most common approach used to identify and quantify analytes from diseased periradicular tissues during root canal treatment, since it is more representative of definitive and active periradicular inflammatory mediator than its transcript expression level. In disease, proteins expressed in an altered manner could be utilized as biomarkers. Biomarker proteins in periradicular tissues have been qualitatively and quantitatively assessed using antibodies (immunoassays and immunostaining) or mass spectrometry-based approaches. Herein, we aim to provide a comprehensive understanding of biomarker proteins identified in clinical studies investigating periradicular lesions and pulp tissue associated with apical periodontitis using proteomics. The high throughput mass spectrometry-based proteomics has the potential to improve the current methods of monitoring inflammation while distinguishing between progressive, stable, and healing lesions for the identification of new diagnostic and therapeutic targets. This method would provide more objective tools to (a) discover biomarkers related to biological processes for better clinical case selection, and (b) determine tissue response to novel therapeutic interventions for more predictable outcomes in endodontic treatment.
Collapse
|
9
|
Abstract
Diarrheal disease is still a major public health concern, as it is still considered an important cause of death in children under five years of age. A few decades ago, the detection of enteropathogenic E. coli was made by detecting the O, H, and K antigens, mostly by agglutination. The recent protocols recommend the molecular methods for diagnosing EPEC, as they can distinguish between typical and atypical EPEC by identifying the presence/absence of specific virulence factors. EPEC are defined as diarrheagenic strains of E. coli that can produce attaching and effacing lesions on the intestinal epithelium while being incapable of producing Shiga toxins and heat-labile or heat-stable enterotoxins. The ability of these strains to produce attaching and effacing lesions enable them to cause localized lesions by attaching tightly to the surface of the intestinal epithelial cells, disrupting the surfaces of the cells, thus leading to the effacement of the microvilli. EPEC are classified on typical and atypical isolates, based on the presence or absence of E. coli adherence factor plasmids. All the EPEC strains are eae positive; typical EPEC strains are eae+, bfpA+, while atypical strains are eae+, bfpA−. No vaccines are currently available to prevent EPEC infections.
Collapse
|
10
|
Shini S, Aland RC, Bryden WL. Avian intestinal ultrastructure changes provide insight into the pathogenesis of enteric diseases and probiotic mode of action. Sci Rep 2021; 11:167. [PMID: 33420315 PMCID: PMC7794591 DOI: 10.1038/s41598-020-80714-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/24/2020] [Indexed: 01/27/2023] Open
Abstract
Epithelial damage and loss of barrier integrity occur following intestinal infections in humans and animals. Gut health was evaluated by electron microscopy in an avian model that exposed birds to subclinical necrotic enteritis (NE) and fed them a diet supplemented with the probiotic Bacillus amyloliquefaciens strain H57 (H57). Scanning electron microscopy of ileal mucosa revealed significant villus damage, including focal erosions of epithelial cells and villous atrophy, while transmission electron microscopy demonstrated severe enterocyte damage and loss of cellular integrity in NE-exposed birds. In particular, mitochondria were morphologically altered, appearing irregular in shape or swollen, and containing electron-lucent regions of matrix and damaged cristae. Apical junctional complexes between adjacent enterocytes were significantly shorter, and the adherens junction was saccular, suggesting loss of epithelial integrity in NE birds. Segmented filamentous bacteria attached to villi, which play an important role in intestinal immunity, were more numerous in birds exposed to NE. The results suggest that mitochondrial damage may be an important initiator of NE pathogenesis, while H57 maintains epithelium and improves the integrity of intestinal mucosa. Potential actions of H57 are discussed that further define the mechanisms responsible for probiotic bacteria’s role in maintaining gut health.
Collapse
Affiliation(s)
- Shaniko Shini
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia.
| | - R Claire Aland
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4071, Australia
| | - Wayne L Bryden
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia
| |
Collapse
|
11
|
Granica S, Vahjen W, Zentek J, Melzig MF, Pawłowska KA, Piwowarski JP. Lythrum salicaria Ellagitannins Stimulate IPEC-J2 Cells Monolayer Formation and Inhibit Enteropathogenic Escherichia coli Growth and Adhesion. JOURNAL OF NATURAL PRODUCTS 2020; 83:3614-3622. [PMID: 33270444 PMCID: PMC7771025 DOI: 10.1021/acs.jnatprod.0c00776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Indexed: 06/12/2023]
Abstract
Lythrum salicaria herb (LSH) was applied in diarrhea therapy since ancient times. Despite empirically referenced therapeutic effects, the bioactivity mechanisms and chemical constituents responsible for pharmacological activity remain not fully resolved. Taking into consideration the historical use of LSH in treatment of diarrhea in humans and farm animals, the aim of the study was to examine in vitro the influence of LSH and its C-glycosylic ellagitannins on processes associated with maintaining intestinal epithelium integrity and enteropathogenic Escherichia coli (EPEC) growth and adhesion. LSH was not only inhibiting EPEC growth in a concentration dependent manner but also its adhesion to IPEC-J2 intestinal epithelial cell monolayers. Inhibitory activity toward EPEC growth was additionally confirmed ex vivo in distal colon samples of postweaning piglets. LSH and its dominating C-glycosylic ellagitannins, castalagin (1), vescalagin (2), and salicarinins A (3) and B (4) were stimulating IPEC-J2 monolayer formation by enhancing claudin 4 production. Parallelly tested gut microbiota metabolites of LSH ellagitannins, urolithin C (5), urolithin A (6), and its glucuronides (7) were inactive. The activities of LSH and the isolated ellagitannins support its purported antidiarrheal properties and indicate potential mechanisms responsible for its beneficial influence on the intestinal epithelium.
Collapse
Affiliation(s)
- Sebastian Granica
- Department
of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw 02-097, Poland
- Centre for Preclinical Studies, Medical
University of Warsaw, Warsaw 02-097, Poland
| | - Wilfried Vahjen
- Institute of Animal Nutrition, Freie Universität
Berlin, Berlin 14195, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität
Berlin, Berlin 14195, Germany
| | - Matthias F. Melzig
- Department of Pharmaceutical Biology, Freie
Universität Berlin, Berlin 14195, Germany
| | - Karolina A. Pawłowska
- Department
of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw 02-097, Poland
- Centre for Preclinical Studies, Medical
University of Warsaw, Warsaw 02-097, Poland
| | - Jakub P. Piwowarski
- Department
of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw 02-097, Poland
- Centre for Preclinical Studies, Medical
University of Warsaw, Warsaw 02-097, Poland
- Institute of Animal Nutrition, Freie Universität
Berlin, Berlin 14195, Germany
- Department of Pharmaceutical Biology, Freie
Universität Berlin, Berlin 14195, Germany
| |
Collapse
|
12
|
Dhanda AS, Yang D, Kooner A, Guttman JA. Distribution of PDLIM1 at actin-rich structures generated by invasive and adherent bacterial pathogens. Anat Rec (Hoboken) 2020; 304:919-938. [PMID: 33022122 DOI: 10.1002/ar.24523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022]
Abstract
The enteric bacterial pathogens Listeria monocytogenes (Listeria) and enteropathogenic Escherichia coli (EPEC) remodel the eukaryotic actin cytoskeleton during their disease processes. Listeria generate slender actin-rich comet/rocket tails to move intracellularly, and later, finger-like membrane protrusions to spread amongst host cells. EPEC remain extracellular, but generate similar actin-rich membranous protrusions (termed pedestals) to move atop the host epithelia. These structures are crucial for disease as diarrheal (and systemic) infections are significantly abrogated during infections with mutant strains that are unable to generate the structures. The current repertoire of host components enriched within these structures is vast and diverse. In this protein catalog, we and others have found that host actin crosslinkers, such as palladin and α-actinin-1, are routinely exploited. To expand on this list, we set out to investigate the distribution of PDLIM1, a scaffolding protein and binding partner of palladin and α-actinin-1, during bacterial infections. We show that PDLIM1 localizes to the site of initial Listeria entry into cells. Following this, PDLIM1 localizes to actin filament clouds surrounding immotile bacteria, and then colocalizes with actin once the comet/rocket tails are generated. Unlike palladin or α-actinin-1, PDLIM1 is maintained within the actin-rich core of membrane protrusions. Conversely, α-actinin-1, but not PDLIM1 (or palladin), is enriched at the membrane invagination that internalizes the Listeria-containing membrane protrusion. We also show that PDLIM1 is a component of the EPEC pedestal core and that its recruitment is dependent on the bacterial effector Tir. Our findings highlight PDLIM1 as another protein present within pathogen-induced actin-rich structures.
Collapse
Affiliation(s)
- Aaron S Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Diana Yang
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Avneen Kooner
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
13
|
Sharba S, Venkatakrishnan V, Padra M, Winther M, Gabl M, Sundqvist M, Wang J, Forsman H, Linden SK. Formyl peptide receptor 2 orchestrates mucosal protection against Citrobacter rodentium infection. Virulence 2020; 10:610-624. [PMID: 31234710 PMCID: PMC6629182 DOI: 10.1080/21505594.2019.1635417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Citrobacter rodentium is an attaching and effacing intestinal murine pathogen which shares similar virulence strategies with the human pathogens enteropathogenic- and enterohemorrhagic Escherichia coli to infect their host. C. rodentium is spontaneously cleared by healthy wild-type (WT) mice whereas mice lacking Muc2 or specific immune regulatory genes demonstrate an impaired ability to combat the pathogen. Here we demonstrate that apical formyl peptide receptor 2 (Fpr2) expression increases in colonic epithelial cells during C. rodentium infection. Using a conventional inoculum dose of C. rodentium, both WT and Fpr2−/− mice were infected and displayed similar signs of disease, although Fpr2−/− mice recovered more slowly than WT mice. However, Fpr2−/− mice exhibited increased susceptibility to C. rodentium colonization in response to low dose infection: 100% of the Fpr2−/− and 30% of the WT mice became colonized and Fpr2−/− mice developed more severe colitis and more C. rodentium were in contact with the colonic epithelial cells. In line with the larger amount of C. rodentium detected in the spleen in Fpr2−/− mice, more C. rodentium and enteropathogenic Escherichia coli translocated across an in vitro mucosal surface to the basolateral compartment following FPR2 inhibitor treatment. Fpr2−/− mice also lacked the striated inner mucus layer that was present in WT mice. Fpr2−/− mice had decreased mucus production and different mucin O-glycosylation in the colon compared to WT mice, which may contribute to their defect inner mucus layer. Thus, Fpr2 contributes to protection against infection and influence mucus production, secretion and organization.
Collapse
Affiliation(s)
- S Sharba
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - V Venkatakrishnan
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Padra
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Winther
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Gabl
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Sundqvist
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - J Wang
- c Cancer and Inflammation Program , National Cancer Institute at Frederick , Frederick , MD , USA
| | - H Forsman
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - S K Linden
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| |
Collapse
|
14
|
Shridhar PB, Patel IR, Gangiredla J, Noll LW, Shi X, Bai J, Nagaraja TG. DNA Microarray-Based Genomic Characterization of the Pathotypes of Escherichia coli O26, O45, O103, O111, and O145 Isolated from Feces of Feedlot Cattle †. J Food Prot 2019; 82:395-404. [PMID: 30794460 DOI: 10.4315/0362-028x.jfp-18-393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Shiga toxin-producing Escherichia coli (STEC) serogroups O26, O45, O103, O111, O121, and O145, referred to as the top six non-O157 serogroups, are responsible for more than 70% of human non-O157 STEC infections in North America. Cattle harbor non-O157 strains in the hindgut and shed them in the feces. The objective of this study was to use the U.S. Food and Drug Administration (FDA) E. coli identification (ECID) DNA microarray to identify the serotype, assess the virulence potential of each, and determine the phylogenetic relationships among five of the six non-O157 E. coli serogroups isolated from feedlot cattle feces. Forty-four strains of STEC, enterohemorrhagic E. coli (EHEC), enteropathogenic E. coli (EPEC), or putative nonpathotype E. coli (NPEC) of cattle origin and five human clinical strains of EHEC were assayed with the FDA-ECID DNA microarray. The cattle strains harbored diverse flagellar genes. The bovine and human strains belonging to serogroups O26, O45, and O103 carried stx1 only, O111 carried both stx1 and stx2, and O145 carried either stx1 or stx2. The strains were also positive for various subtypes of intimin and other adhesins (IrgA homologue adhesin, long polar fimbriae, mannose-specific adhesin, and curli). Both human and cattle strains were positive for LEE-encoded type III secretory system genes and non-LEE-encoded effector genes. SplitsTree4, a program used to determine the phylogenetic relationship among the strains, revealed that the strains within each serogroup clustered according to their pathotype. In addition to genes encoding Shiga toxins, bovine non-O157 E. coli strains possessed other major virulence genes, including those for adhesins, type III secretory system proteins, and plasmid-borne virulence genes, similar to human clinical strains. Because virulence factors encoded by these genes are involved in the pathogenesis of various pathotypes of E. coli, the bovine non-O157 strains could cause human illness. The FDA-ECID DNA microarray assay rapidly provided a profile of the virulence genes for assessment of the virulence potential of each strain.
Collapse
Affiliation(s)
- Pragathi B Shridhar
- 1 Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas 66506
| | - Isha R Patel
- 2 U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, Division of Molecular Biology, Laurel, Maryland 20708, USA
| | - Jayanthi Gangiredla
- 2 U.S. Food and Drug Administration, Center for Food Safety and Applied Nutrition, Division of Molecular Biology, Laurel, Maryland 20708, USA
| | - Lance W Noll
- 1 Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas 66506
| | - Xiaorong Shi
- 1 Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas 66506
| | - Jianfa Bai
- 3 Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, Kansas 66506
| | - T G Nagaraja
- 1 Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas 66506
| |
Collapse
|
15
|
Chua MD, Walker BD, Jin JP, Guttman JA. Calponins Are Recruited to Actin-Rich Structures Generated by Pathogenic Escherichia coli, Listeria, and Salmonella. Anat Rec (Hoboken) 2018; 301:2103-2111. [PMID: 30312538 DOI: 10.1002/ar.23956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/12/2023]
Abstract
The ingestion of enteropathogenic Escherichia coli (EPEC), Listeria monocytogenes, or Salmonella enterica serovar Typhimurium leads to their colonization of the intestinal lumen, which ultimately causes an array of ailments ranging from diarrhea to bacteremia. Once in the intestines, these microbes generate various actin-rich structures to attach, invade, or move within the host intestinal epithelial cells. Although an assortment of actin-associated proteins has been identified to varying degrees at these structures, the localization of many actin stabilizing proteins have yet to be analyzed. Here, we examined the recruitment of the actin-associated proteins, calponin 1 and 2 at EPEC pedestals, L. monocytogenes actin clouds, comet tails and listeriopods, and S. Typhimurium membrane ruffles. In other systems, calponins are known to bind to and stabilize actin filaments. In EPEC pedestals, calponin 1 was recruited uniformly throughout the structures while calponin 2 was enriched at the apical tip. During L. monocytogenes infections, calponin 1 was found through all the actin-rich structures generated by the bacteria, while calponin 2 was only present within actin-rich structures formed by L. monocytogenes near the host cell membrane. Finally, both calponins were found within S. Typhimurium-generated membrane ruffles. Taken together, we have shown that although calponin 1 is recruited to actin-rich structures formed by the three bacteria, calponin 2 is specifically recruited to only membrane-bound actin-rich structures formed by the bacteria. Thus, our findings suggest that calponin 2 is a novel marker for membrane-bound actin structures formed by pathogenic bacteria. Anat Rec, 301:2103-2111, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael Dominic Chua
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Julian A Guttman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
16
|
Poudineh M, Wang Z, Labib M, Ahmadi M, Zhang L, Das J, Ahmed S, Angers S, Kelley SO. Three-Dimensional Nanostructured Architectures Enable Efficient Neural Differentiation of Mesenchymal Stem Cells via Mechanotransduction. NANO LETTERS 2018; 18:7188-7193. [PMID: 30335391 DOI: 10.1021/acs.nanolett.8b03313] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Cell morphology and geometry affect cellular processes such as stem cell differentiation, suggesting that these parameters serve as fundamental regulators of biological processes within the cell. Hierarchical architectures featuring micro- and nanotopographical features therefore offer programmable systems for stem cell differentiation. However, a limited number of studies have explored the effects of hierarchical architectures due to the complexity of fabricating systems with rationally tunable micro- and nanostructuring. Here, we report three-dimensional (3D) nanostructured microarchitectures that efficiently regulate the fate of human mesenchymal stem cells (hMSCs). These nanostructured architectures strongly promote cell alignment and efficient neurogenic differentiation where over 85% of hMSCs express microtubule-associated protein 2 (MAP2), a mature neural marker, after 7 days of culture on the nanostructured surface. Remarkably, we found that the surface morphology of nanostructured surface is a key factor that promotes neurogenesis and that highly spiky structures promote more efficient neuronal differentiation. Immunostaining and gene expression profiling revealed significant upregulation of neuronal markers compared to unpatterned surfaces. These findings suggest that the 3D nanostructured microarchitectures can play a critical role in defining stem cell behavior.
Collapse
Affiliation(s)
- Mahla Poudineh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Zongjie Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Moloud Ahmadi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Libing Zhang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Jagotamoy Das
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Sharif Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , M5S 3M2 , Canada
- Institute for Biomaterials and Biomedical Engineering , University of Toronto , Toronto , M5S 3M2 , Canada
- Department of Biochemistry, Faculty of Medicine , University of Toronto , Toronto , M5S 1A8 , Canada
| |
Collapse
|
17
|
Cavalcante PA, Prata MMG, Medeiros PHQS, Alves da Silva AV, Quetz JS, Reyes MAV, Rodrigues TS, Santos AKS, Ribeiro SA, Veras HN, Bona MD, Amaral MSMG, Rodrigues FAP, Lima IFN, Havt A, Lima AAM. Intestinal cell migration damage induced by enteropathogenic Escherichia coli strains. ACTA ACUST UNITED AC 2018; 51:e7423. [PMID: 30066727 PMCID: PMC6065879 DOI: 10.1590/1414-431x20187423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/24/2018] [Indexed: 01/27/2023]
Abstract
Epithelial cell migration is an essential response to enteric pathogens such as enteropathogenic Escherichia coli (EPEC). This study aimed to investigate the effects of EPEC infection on intestinal epithelial cell migration in vitro, as well as the involvement of type III secretion system (T3SS) and Rho GTPases. Crypt intestinal epithelial cells (IEC-6) were infected with EPEC strains (E2348/69, ΔescF, and the LDI001 strain isolated from a malnourished Brazilian child) and commensal E. coli HS. Wound migration and cell death assays were performed at different time-points. Transcription and expression of Rho GTPases were evaluated using real-time PCR and western blotting. Overall, EPEC E2348/69 reduced migration and increased apoptosis and necrosis levels compared to EPEC LDI001 and E. coli HS strains. Moreover, EPEC LDI001 impaired cell migration at a higher level than E. coli HS and increased necrosis after 24 hours compared to the control group. The different profiles of virulence genes between the two wild-type EPEC strains, characterized by the absence of espL and nleE genes in the LDI001, might explain the phenotypic results, playing significant roles on cell migration impairment and cell death-related events. Moreover, the type III secretion system is determinant for the inhibition of intestinal epithelial cell migration by EPEC 2348/69, as its deletion prevented the effect. Active Rac1 concentrations were increased in E2348/69 and LDI001-infected cells, while the T3SS-deficient strain did not demonstrate this activation. This study contributes with valuable insight to characterize the mechanisms involved in the impairment of intestinal cell migration induced by EPEC.
Collapse
Affiliation(s)
- P A Cavalcante
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M M G Prata
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - P H Q S Medeiros
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A V Alves da Silva
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - J S Quetz
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M A V Reyes
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T S Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A K S Santos
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S A Ribeiro
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - H N Veras
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M D Bona
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - M S M G Amaral
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - F A P Rodrigues
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - I F N Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A Havt
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A A M Lima
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
18
|
Martínez de Castro DL, García-Gómez BI, Gómez I, Bravo A, Soberón M. Identification of Bacillus thuringiensis Cry1AbMod binding-proteins from Spodoptera frugiperda. Peptides 2017; 98:99-105. [PMID: 28958733 DOI: 10.1016/j.peptides.2017.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/06/2017] [Accepted: 09/22/2017] [Indexed: 12/12/2022]
Abstract
Bacillus thuringiensis Cry toxins are currently used for pest control in transgenic crops but evolution of resistance by the insect pests threatens the use of this technology. The Cry1AbMod toxin was engineered to lack the alpha helix-1 of the parental Cry1Ab toxin and was shown to counter resistance to Cry1Ab and Cry1Ac toxins in different insect species including the fall armyworm Spodoptera frugiperda. In addition, Cry1AbMod showed enhanced toxicity to Cry1Ab-susceptible S. frugiperda populations. To gain insights into the mechanisms of this Cry1AbMod-enhanced toxicity, we isolated the Cry1AbMod toxin binding proteins from S. frugiperda brush border membrane vesicles (BBMV), which were identified by pull-down assay and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The LC-MS/MS results indicated that Cry1AbMod toxin could bind to four classes of aminopeptidase (N1, N3, N4 y N5) and actin, with the highest amino acid sequence coverage acquired for APN 1 and APN4. In addition to these proteins, we found other proteins not previously described as Cry toxin binding proteins. This is the first report that suggests the interaction between Cry1AbMod and APN in S. frugiperda.
Collapse
Affiliation(s)
- Diana L Martínez de Castro
- Instituto de Biotecnología, Universidad Nacional Autónoma de México. Apdo. Postal 510-3, Cuernavaca, 62250 Morelos, Mexico
| | - Blanca I García-Gómez
- Instituto de Biotecnología, Universidad Nacional Autónoma de México. Apdo. Postal 510-3, Cuernavaca, 62250 Morelos, Mexico
| | - Isabel Gómez
- Instituto de Biotecnología, Universidad Nacional Autónoma de México. Apdo. Postal 510-3, Cuernavaca, 62250 Morelos, Mexico
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México. Apdo. Postal 510-3, Cuernavaca, 62250 Morelos, Mexico
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México. Apdo. Postal 510-3, Cuernavaca, 62250 Morelos, Mexico.
| |
Collapse
|
19
|
Escherichia albertii, a novel human enteropathogen, colonizes rat enterocytes and translocates to extra-intestinal sites. PLoS One 2017; 12:e0171385. [PMID: 28178312 PMCID: PMC5298312 DOI: 10.1371/journal.pone.0171385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/18/2017] [Indexed: 01/01/2023] Open
Abstract
Diarrhea is the second leading cause of death of children up to five years old in the developing countries. Among the etiological diarrheal agents are atypical enteropathogenic Escherichia coli (aEPEC), one of the diarrheagenic E. coli pathotypes that affects children and adults, even in developed countries. Currently, genotypic and biochemical approaches have helped to demonstrate that some strains classified as aEPEC are actually E. albertii, a recently recognized human enteropathogen. Studies on particular strains are necessary to explore their virulence potential in order to further understand the underlying mechanisms of E. albertii infections. Here we demonstrated for the first time that infection of fragments of rat intestinal mucosa is a useful tool to study the initial steps of E. albertii colonization. We also observed that an E. albertii strain can translocate from the intestinal lumen to Mesenteric Lymph Nodes and liver in a rat model. Based on our finding of bacterial translocation, we investigated how E. albertii might cross the intestinal epithelium by performing infections of M-like cells in vitro to identify the potential in vivo translocation route. Altogether, our approaches allowed us to draft a general E. albertii infection route from the colonization till the bacterial spreading in vivo.
Collapse
|
20
|
Janik K, Schlink K. Unravelling the Function of a Bacterial Effector from a Non-cultivable Plant Pathogen Using a Yeast Two-hybrid Screen. J Vis Exp 2017. [PMID: 28190069 PMCID: PMC5352286 DOI: 10.3791/55150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Unravelling the molecular mechanisms of disease manifestations is important to understand pathologies and symptom development in plant science. Bacteria have evolved different strategies to manipulate their host metabolism for their own benefit. This bacterial manipulation is often coupled with severe symptom development or the death of the affected plants. Determining the specific bacterial molecules responsible for the host manipulation has become an important field in microbiological research. After the identification of these bacterial molecules, called "effectors," it is important to elucidate their function. A straightforward approach to determine the function of an effector is to identify its proteinaceous binding partner in its natural host via a yeast two-hybrid (Y2H) screen. Normally the host harbors numerous potential binding partners that cannot be predicted sufficiently by any in silico algorithm. It is thus the best choice to perform a screen with the hypothetical effector against a whole library of expressed host proteins. It is especially challenging if the causative agent is uncultivable like phytoplasma. This protocol provides step-by-step instructions for DNA purification from a phytoplasma-infected woody host plant, the amplification of the potential effector, and the subsequent identification of the plant's molecular interaction partner with a Y2H screen. Even though Y2H screens are commonly used, there is a trend to outsource this technique to biotech companies that offer the Y2H service at a cost. This protocol provides instructions on how to perform a Y2H in any decently equipped molecular biology laboratory using standard lab techniques.
Collapse
Affiliation(s)
- Katrin Janik
- Department of Molecular Biology - Functional Genomics, Laimburg Research Centre;
| | - Katja Schlink
- Department of Molecular Biology - Functional Genomics, Laimburg Research Centre
| |
Collapse
|
21
|
Mittal R, Lisi CV, Kumari H, Grati M, Blackwelder P, Yan D, Jain C, Mathee K, Weckwerth PH, Liu XZ. Otopathogenic Pseudomonas aeruginosa Enters and Survives Inside Macrophages. Front Microbiol 2016; 7:1828. [PMID: 27917157 PMCID: PMC5114284 DOI: 10.3389/fmicb.2016.01828] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Otitis media (OM) is a broad term describing a group of infectious and inflammatory disorders of the middle ear. Despite antibiotic therapy, acute OM can progress to chronic suppurative otitis media (CSOM) characterized by ear drum perforation and purulent discharge. Pseudomonas aeruginosa is the most common pathogen associated with CSOM. Although, macrophages play an important role in innate immune responses but their role in the pathogenesis of P. aeruginosa-induced CSOM is not known. The objective of this study is to examine the interaction of P. aeruginosa with primary macrophages. We observed that P. aeruginosa enters and multiplies inside human and mouse primary macrophages. This bacterial entry in macrophages requires both microtubule and actin dependent processes. Transmission electron microscopy demonstrated that P. aeruginosa was present in membrane bound vesicles inside macrophages. Interestingly, deletion of oprF expression in P. aeruginosa abrogates its ability to survive inside macrophages. Our results suggest that otopathogenic P. aeruginosa entry and survival inside macrophages is OprF-dependent. The survival of bacteria inside macrophages will lead to evasion of killing and this lack of pathogen clearance by phagocytes contributes to the persistence of infection in CSOM. Understanding host-pathogen interaction will provide novel avenues to design effective treatment modalities against OM.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Christopher V Lisi
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Hansi Kumari
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami FL, USA
| | - M'hamed Grati
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Patricia Blackwelder
- Chemistry Department, Center for Advanced Microscopy, University of Miami, Coral GablesFL, USA; Rosenstiel School of Marine and Atmospheric Science, University of Miami, Key BiscayneFL, USA
| | - Denise Yan
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Chaitanya Jain
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami FL, USA
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, MiamiFL, USA; Global Health Consortium and Biomolecular Science Institute, Florida International University, MiamiFL, USA
| | - Paulo H Weckwerth
- Health Sciences Department, University of Sagrado Coração Bauru, Brazil
| | - Xue Z Liu
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami FL, USA
| |
Collapse
|
22
|
Ugalde-Silva P, Gonzalez-Lugo O, Navarro-Garcia F. Tight Junction Disruption Induced by Type 3 Secretion System Effectors Injected by Enteropathogenic and Enterohemorrhagic Escherichia coli. Front Cell Infect Microbiol 2016; 6:87. [PMID: 27606286 PMCID: PMC4995211 DOI: 10.3389/fcimb.2016.00087] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/10/2016] [Indexed: 12/18/2022] Open
Abstract
The intestinal epithelium consists of a single cell layer, which is a critical selectively permeable barrier to both absorb nutrients and avoid the entry of potentially harmful entities, including microorganisms. Epithelial cells are held together by the apical junctional complexes, consisting of adherens junctions, and tight junctions (TJs), and by underlying desmosomes. TJs lay in the apical domain of epithelial cells and are mainly composed by transmembrane proteins such as occludin, claudins, JAMs, and tricellulin, that are associated with the cytoplasmic plaque formed by proteins from the MAGUK family, such as ZO-1/2/3, connecting TJ to the actin cytoskeleton, and cingulin and paracingulin connecting TJ to the microtubule network. Extracellular bacteria such as EPEC and EHEC living in the intestinal lumen inject effectors proteins directly from the bacterial cytoplasm to the host cell cytoplasm, where they play a relevant role in the manipulation of the eukaryotic cell functions by modifying or blocking cell signaling pathways. TJ integrity depends on various cell functions such as actin cytoskeleton, microtubule network for vesicular trafficking, membrane integrity, inflammation, and cell survival. EPEC and EHEC effectors target most of these functions. Effectors encoded inside or outside of locus of enterocyte effacement (LEE) disrupt the TJ strands. EPEC and EHEC exploit the TJ dynamics to open this structure, for causing diarrhea. EPEC and EHEC secrete effectors that mimic host proteins to manipulate the signaling pathways, including those related to TJ dynamics. In this review, we focus on the known mechanisms exploited by EPEC and EHEC effectors for causing TJ disruption.
Collapse
Affiliation(s)
- Paul Ugalde-Silva
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| | - Octavio Gonzalez-Lugo
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| |
Collapse
|
23
|
Wang X, Li J, Chen S, Shen X, Yang X, Teng Y, Deng L, Wang Y, Chen J, Wang X, Huang N. Knockdown of HMGN2 increases the internalization of Klebsiella pneumoniae by respiratory epithelial cells through the regulation of α5β1 integrin expression. Int J Mol Med 2016; 38:737-46. [PMID: 27460641 PMCID: PMC4990306 DOI: 10.3892/ijmm.2016.2690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 07/11/2016] [Indexed: 01/13/2023] Open
Abstract
Integrin receptors, a large family of adhesion receptors, are involved in the attachment of Klebsiella pneumoniae to respiratory epithelial cells, and subsequently cause the internalization of K. pneumoniae by host cells. Although a number of molecules have been reported to regulate the expression and activity of integrin receptors in respiratory epithelial cells, the specific underlying molecular mechanisms remain largely unknown. High mobility group nucleosomal binding domain 2 (HMGN2), a non-histone nuclear protein, is present in eukaryotic cells as a ubiquitous nuclear protein. Our previous studies have demonstrated that HMGN2 affects chromatin function and modulates the expression of antibacterial peptide in A549 cells exposed to lipopolysaccharide, which indicates the critical role of HMGN2 in innate immune responses. In addition, our cDNA microarray analysis suggested that HMGN2 knockdown induced the enhanced expression of α5β1 integrin in A549 cells. Therefore, we hypothesized that intercellular HMGN2 may mediate the internalization of K. pneumoniae by altering the expression of α5β1 integrin. Using the A549 cell line, we demonstrated that HMGN2 knockdown induced the increased expression of α5β1 integrin on cell membranes, which resulted in a significant increase in K. pneumoniae internalization. Further results revealed that HMGN2 silencing induced the expression of talin and the activation of α5β1 integrin, which led to actin polymerization following the phosphorylation of FAK and Src. This study suggests a possible therapeutic application for bacterial internalization by targeting HMGN2 in order to treat K. pneumoniae infection.
Collapse
Affiliation(s)
- Xinyuan Wang
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jingyu Li
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shanze Chen
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaofei Shen
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaolong Yang
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Teng
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Luxia Deng
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi Wang
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Junli Chen
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaoying Wang
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ning Huang
- Department of Pathophysiology, Research Unit of Infection and Immunity, West China College of Basic and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
24
|
Anti-infective activities of lactobacillus strains in the human intestinal microbiota: from probiotics to gastrointestinal anti-infectious biotherapeutic agents. Clin Microbiol Rev 2016; 27:167-99. [PMID: 24696432 DOI: 10.1128/cmr.00080-13] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A vast and diverse array of microbial species displaying great phylogenic, genomic, and metabolic diversity have colonized the gastrointestinal tract. Resident microbes play a beneficial role by regulating the intestinal immune system, stimulating the maturation of host tissues, and playing a variety of roles in nutrition and in host resistance to gastric and enteric bacterial pathogens. The mechanisms by which the resident microbial species combat gastrointestinal pathogens are complex and include competitive metabolic interactions and the production of antimicrobial molecules. The human intestinal microbiota is a source from which Lactobacillus probiotic strains have often been isolated. Only six probiotic Lactobacillus strains isolated from human intestinal microbiota, i.e., L. rhamnosus GG, L. casei Shirota YIT9029, L. casei DN-114 001, L. johnsonii NCC 533, L. acidophilus LB, and L. reuteri DSM 17938, have been well characterized with regard to their potential antimicrobial effects against the major gastric and enteric bacterial pathogens and rotavirus. In this review, we describe the current knowledge concerning the experimental antibacterial activities, including antibiotic-like and cell-regulating activities, and therapeutic effects demonstrated in well-conducted, placebo-controlled, randomized clinical trials of these probiotic Lactobacillus strains. What is known about the antimicrobial activities supported by the molecules secreted by such probiotic Lactobacillus strains suggests that they constitute a promising new source for the development of innovative anti-infectious agents that act luminally and intracellularly in the gastrointestinal tract.
Collapse
|
25
|
Escherichia coli O104:H4 Pathogenesis: an Enteroaggregative E. coli/Shiga Toxin-Producing E. coli Explosive Cocktail of High Virulence. Microbiol Spectr 2016; 2. [PMID: 26104460 DOI: 10.1128/microbiolspec.ehec-0008-2013] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A major outbreak caused by Escherichia coli of serotype O104:H4 spread throughout Europe in 2011. This large outbreak was caused by an unusual strain that is most similar to enteroaggregative E. coli (EAEC) of serotype O104:H4. A significant difference, however, is the presence of a prophage encoding the Shiga toxin, which is characteristic of enterohemorrhagic E. coli (EHEC) strains. This combination of genomic features, associating characteristics from both EAEC and EHEC, represents a new pathotype. The 2011 E. coli O104:H4 outbreak of hemorrhagic diarrhea in Germany is an example of the explosive cocktail of high virulence and resistance that can emerge in this species. A total of 46 deaths, 782 cases of hemolytic-uremic syndrome, and 3,128 cases of acute gastroenteritis were attributed to this new clone of EAEC/EHEC. In addition, recent identification in France of similar O104:H4 clones exhibiting the same virulence factors suggests that the EHEC O104:H4 pathogen has become endemically established in Europe after the end of the outbreak. EAEC strains of serotype O104:H4 contain a large set of virulence-associated genes regulated by the AggR transcription factor. They include, among other factors, the pAA plasmid genes encoding the aggregative adherence fimbriae, which anchor the bacterium to the intestinal mucosa (stacked-brick adherence pattern on epithelial cells). Furthermore, sequencing studies showed that horizontal genetic exchange allowed for the emergence of the highly virulent Shiga toxin-producing EAEC O104:H4 strain that caused the German outbreak. This article discusses the role these virulence factors could have in EAEC/EHEC O104:H4 pathogenesis.
Collapse
|
26
|
Jensen HH, Pedersen HN, Stenkjær E, Pedersen GA, Login FH, Nejsum LN. Tir Is Essential for the Recruitment of Tks5 to Enteropathogenic Escherichia coli Pedestals. PLoS One 2015; 10:e0141871. [PMID: 26536015 PMCID: PMC4633291 DOI: 10.1371/journal.pone.0141871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a bacterial pathogen that infects the epithelial lining of the small intestine and causes diarrhea. Upon attachment to the intestinal epithelium, EPEC uses a Type III Secretion System to inject its own high affinity receptor Translocated intimin receptor (Tir) into the host cell. Tir facilitates tight adhesion and recruitment of actin-regulating proteins leading to formation of an actin pedestal beneath the infecting bacterium. The pedestal has several similarities with podosomes, which are basolateral actin-rich extensions found in some migrating animal cells. Formation of podosomes is dependent upon the early podosome-specific scavenger protein Tks5, which is involved in actin recruitment. Although Tks5 is expressed in epithelial cells, and podosomes and EPEC pedestals share many components in their structure and mechanism of formation, the potential role of Tks5 in EPEC infections has not been studied. The aim of this study was to determine the subcellular localization of Tks5 in epithelial cells and to investigate if Tks5 is recruited to the EPEC pedestal. In an epithelial MDCK cell line stably expressing Tks5-EGFP, Tks5 localized to actin bundles. Upon infection, EPEC recruited Tks5-EGFP. Tir, but not Tir phosphorylation was essential for the recruitment. Time-lapse microscopy revealed that Tks5-EGFP was recruited instantly upon EPEC attachment to host cells, simultaneously with actin and N-WASp. EPEC infection of cells expressing a ΔPX-Tks5 deletion version of Tks5 showed that EPEC was able to both infect and form pedestals when the PX domain was deleted from Tks5. Future investigations will clarify the role of Tks5 in EPEC infection and pedestal formation.
Collapse
Affiliation(s)
- Helene H. Jensen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Hans N. Pedersen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Eva Stenkjær
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Gitte A. Pedersen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Frédéric H. Login
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Lene N. Nejsum
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
27
|
Deligianni DD. MWCNTs enhance hBMSCs spreading but delay their proliferation in the direction of differentiation acceleration. Cell Adh Migr 2015; 8:404-17. [PMID: 25482637 DOI: 10.4161/19336918.2014.969993] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Investigating the ability of films of pristine multiwalled nanotubes (MWCNTs) to influence human mesenchymal stem cells' proliferation, morphology, and differentiation into osteoblasts, we concluded to the following: A. MWCNTs delay the proliferation of hBMS cells but increase their differentiation. The enhancement of the differentiation markers could be a result of decreased proliferation and maturation of the extracellular matrix B. Cell spread on MWCNTs toward a polygonal shape with many thin filopodia to attach to the surfaces. Spreading may be critical in supporting osteogenic differentiation in pre-osteoblastic progenitors, being related with cytoskeletal tension. C. hBMS cells prefer MWCNTs than tissue plastic to attach and grow, being non-toxic to these cells. MWCNTs can be regarded as osteoinductive biomaterial topographies for bone regenerative engineering.
Collapse
Affiliation(s)
- Despina D Deligianni
- a Department of Mechanical Engineering & Aeronautics ; University of Patras ; Rion , Greece
| |
Collapse
|
28
|
Pathogenesis of human diffusely adhering Escherichia coli expressing Afa/Dr adhesins (Afa/Dr DAEC): current insights and future challenges. Clin Microbiol Rev 2015; 27:823-69. [PMID: 25278576 DOI: 10.1128/cmr.00036-14] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenicity and clinical pertinence of diffusely adhering Escherichia coli expressing the Afa/Dr adhesins (Afa/Dr DAEC) in urinary tract infections (UTIs) and pregnancy complications are well established. In contrast, the implication of intestinal Afa/Dr DAEC in diarrhea is still under debate. These strains are age dependently involved in diarrhea in children, are apparently not involved in diarrhea in adults, and can also be asymptomatic intestinal microbiota strains in children and adult. This comprehensive review analyzes the epidemiology and diagnosis and highlights recent progress which has improved the understanding of Afa/Dr DAEC pathogenesis. Here, I summarize the roles of Afa/Dr DAEC virulence factors, including Afa/Dr adhesins, flagella, Sat toxin, and pks island products, in the development of specific mechanisms of pathogenicity. In intestinal epithelial polarized cells, the Afa/Dr adhesins trigger cell membrane receptor clustering and activation of the linked cell signaling pathways, promote structural and functional cell lesions and injuries in intestinal barrier, induce proinflammatory responses, create angiogenesis, instigate epithelial-mesenchymal transition-like events, and lead to pks-dependent DNA damage. UTI-associated Afa/Dr DAEC strains, following adhesin-membrane receptor cell interactions and activation of associated lipid raft-dependent cell signaling pathways, internalize in a microtubule-dependent manner within urinary tract epithelial cells, develop a particular intracellular lifestyle, and trigger a toxin-dependent cell detachment. In response to Afa/Dr DAEC infection, the host epithelial cells generate antibacterial defense responses. Finally, I discuss a hypothetical role of intestinal Afa/Dr DAEC strains that can act as "silent pathogens" with the capacity to emerge as "pathobionts" for the development of inflammatory bowel disease and intestinal carcinogenesis.
Collapse
|
29
|
Nishimura M, Fujii T, Hiyoshi H, Makino F, Inoue H, Motooka D, Kodama T, Ohkubo T, Kobayashi Y, Nakamura S, Namba K, Iida T. A repeat unit of Vibrio diarrheal T3S effector subverts cytoskeletal actin homeostasis via binding to interstrand region of actin filaments. Sci Rep 2015; 5:10870. [PMID: 26039684 PMCID: PMC4650670 DOI: 10.1038/srep10870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/05/2015] [Indexed: 11/09/2022] Open
Abstract
A novel bacterial type III secretion effector, VopV, from the enteric pathogen Vibrio parahaemolyticus has been identified as a key factor in pathogenicity due to its interaction with cytoskeletal actin. One of the repeat units in the long repetitive region of VopV, named VopVrep1, functions as an actin-binding module. Despite its importance in pathogenesis, the manner in which the effector binds to actin and the subsequent effects on actin dynamics remain unclear. Here, we report the molecular basis of the VopVrep1/actin interaction. VopVrep1 exists as an unstructured protein in solution but potently and specifically binds filamentous actin (F-actin) and not globular actin (G-actin). The F-actin/VopVrep1 complex was directly visualized at 9.6-Å resolution using electron cryomicroscopy (cryoEM) and helical image reconstitution. The density map revealed the binding site of VopVrep1 at the interface between two actin strands, which is close to the binding site of the bicyclic heptapeptide toxin phalloidin. Consistent with this observation, VopVrep1 alone prevented the depolymerization of F-actin. Overall, VopVrep1 demonstrated unique characteristics in comparison to known actin-binding proteins, but was relatively similar to phalloidin. The phalloidin-like behavior, targeting the interstrand region of actin filaments to stabilize the filament structure, likely contributes to the pathogenicity of V. parahaemolyticus.
Collapse
Affiliation(s)
- Mitsuhiro Nishimura
- 1] Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Takashi Fujii
- 1] Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Riken Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirotaka Hiyoshi
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Fumiaki Makino
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hajime Inoue
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Daisuke Motooka
- 1] Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka Suita, Osaka 565-0871, Japan
| | - Toshio Kodama
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadayasu Ohkubo
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka Suita, Osaka 565-0871, Japan
| | - Yuji Kobayashi
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Shota Nakamura
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keiichi Namba
- 1] Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Riken Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuya Iida
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
30
|
Scholz R, Imami K, Scott NE, Trimble WS, Foster LJ, Finlay BB. Novel Host Proteins and Signaling Pathways in Enteropathogenic E. coli Pathogenesis Identified by Global Phosphoproteome Analysis. Mol Cell Proteomics 2015; 14:1927-45. [PMID: 25944883 DOI: 10.1074/mcp.m114.046847] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system (T3SS) to directly translocate effector proteins into host cells where they play a pivotal role in subverting host cell signaling needed for disease. However, our knowledge of how EPEC affects host protein phosphorylation is limited to a few individual protein studies. We employed a quantitative proteomics approach to globally map alterations in the host phosphoproteome during EPEC infection. By characterizing host phosphorylation events at various time points throughout infection, we examined how EPEC dynamically impacts the host phosphoproteome over time. This experimental setup also enabled identification of T3SS-dependent and -independent changes in host phosphorylation. Specifically, T3SS-regulated events affected various cellular processes that are known EPEC targets, including cytoskeletal organization, immune signaling, and intracellular trafficking. However, the involvement of phosphorylation in these events has thus far been poorly studied. We confirmed the MAPK family as an established key host player, showed its central role in signal transduction during EPEC infection, and extended the repertoire of known signaling hubs with previously unrecognized proteins, including TPD52, CIN85, EPHA2, and HSP27. We identified altered phosphorylation of known EPEC targets, such as cofilin, where the involvement of phosphorylation has so far been undefined, thus providing novel mechanistic insights into the roles of these proteins in EPEC infection. An overlap of regulated proteins, especially those that are cytoskeleton-associated, was observed when compared with the phosphoproteome of Shigella-infected cells. We determined the biological relevance of the phosphorylation of a novel protein in EPEC pathogenesis, septin-9 (SEPT9). Both siRNA knockdown and a phosphorylation-impaired SEPT9 mutant decreased bacterial adherence and EPEC-mediated cell death. In contrast, a phosphorylation-mimicking SEPT9 mutant rescued these effects. Collectively, this study provides the first global analysis of phosphorylation-mediated processes during infection with an extracellular, diarrheagenic bacterial pathogen.
Collapse
Affiliation(s)
| | - Koshi Imami
- §Centre for High-Throughput Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Nichollas E Scott
- §Centre for High-Throughput Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - William S Trimble
- ¶Cell Biology Program, Hospital for Sick Children and ‖Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Leonard J Foster
- §Centre for High-Throughput Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada, **Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada, and
| | - B Brett Finlay
- From the ‡Michael Smith Laboratories and **Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada, and ¶¶Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
31
|
Antisecretory factor peptide AF-16 inhibits the secreted autotransporter toxin-stimulated transcellular and paracellular passages of fluid in cultured human enterocyte-like cells. Infect Immun 2014; 83:907-22. [PMID: 25534938 DOI: 10.1128/iai.02759-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Both the endogenous antisecretory factor (AF) protein and peptide AF-16, which has a sequence that matches that of the active N-terminal region of AF, inhibit the increase in the epithelial transport of fluid and electrolytes induced by bacterial toxins in animal and ex vivo models. We conducted a study to investigate the inhibitory effect of peptide AF-16 against the increase of transcellular passage and paracellular permeability promoted by the secreted autotransporter toxin (Sat) in a cultured cellular model of the human intestinal epithelial barrier. Peptide AF-16 produced a concentration-dependent inhibition of the Sat-induced increase in the formation of fluid domes, in the mucosal-to-serosal passage of D-[1-(14)C]mannitol, and in the rearrangements in the distribution and protein expression of the tight junction (TJ)-associated proteins ZO-1 and occludin in cultured human enterocyte-like Caco-2/TC7 cell monolayers. In addition, we show that peptide AF-16 also inhibits the cholera toxin-induced increase of transcellular passage and the Clostridium difficile toxin-induced effects on paracellular permeability and TJ protein organization in Caco-2/TC7 cell monolayers. Treatment of cell monolayers by the lipid raft disorganizer methyl-β-cyclodextrin abolished the inhibitory activity of peptide AF-16 at the transcellular passage level and did not modify the effect of the peptide at the paracellular level.
Collapse
|
32
|
Ramírez R, Gómez FA, Marshall SH. The infection process of Piscirickettsia salmonis in fish macrophages is dependent upon interaction with host-cell clathrin and actin. FEMS Microbiol Lett 2014; 362:1-8. [PMID: 25790493 DOI: 10.1093/femsle/fnu012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Piscirickettsia salmonis is an aggressive fish pathogen that causes Piscirickettsiosis, a systemic disease that threatens the sustainability of salmon production in Chile. To date, the infection strategies of this bacterium are poorly characterized, a Dot/Icm Type IV Secretion System homolog for intracellular multiplication and survival in macrophages is suggested. Since an invading pathogen and its host develop a complex interaction in which the pathogen strives to survive and replicate, while the host tries to eliminate infected cells and the invading pathogen, we decided to evaluate how the bacterium enters macrophages, its preferred target in vivo, and to follow its fate while struggling with its host using actin cytoskeleton as a molecular marker. We were able to demonstrate that clathrin is required for internalization and that actin cytoskeleton plays a demonstrative role throughout the infective process. Indeed, unlike other fish pathogens, P. salmonis fully exploits the actin monomers both from the disorganized cytoskeleton and an apparently pathogen-induced de novo synthesis of actin, generating tridimensional vacuoles that are increasingly detected at later stages of infection. We expect our results to contribute to a better understanding of the pathogenesis of this important fish pathogen.
Collapse
Affiliation(s)
- Ramón Ramírez
- Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Universidad 330, Curauma, Valparaíso, Chile Fraunhofer Chile Research Foundation, Center For Systems Biotechnology, Avenida M. Sánchez Fontecilla 310, Piso 14. Las Condes Santiago, Chile
| | - Fernando A Gómez
- Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Universidad 330, Curauma, Valparaíso, Chile
| | - Sergio H Marshall
- Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Avenida Universidad 330, Curauma, Valparaíso, Chile NBC, Núcleo de Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Avenida Universidad 330, Curauma, Valparaíso, Chile Fraunhofer Chile Research Foundation, Center For Systems Biotechnology, Avenida M. Sánchez Fontecilla 310, Piso 14. Las Condes Santiago, Chile
| |
Collapse
|
33
|
Via A, Uyar B, Brun C, Zanzoni A. How pathogens use linear motifs to perturb host cell networks. Trends Biochem Sci 2014; 40:36-48. [PMID: 25475989 DOI: 10.1016/j.tibs.2014.11.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 12/31/2022]
Abstract
Molecular mimicry is one of the powerful stratagems that pathogens employ to colonise their hosts and take advantage of host cell functions to guarantee their replication and dissemination. In particular, several viruses have evolved the ability to interact with host cell components through protein short linear motifs (SLiMs) that mimic host SLiMs, thus facilitating their internalisation and the manipulation of a wide range of cellular networks. Here we present convincing evidence from the literature that motif mimicry also represents an effective, widespread hijacking strategy in prokaryotic and eukaryotic parasites. Further insights into host motif mimicry would be of great help in the elucidation of the molecular mechanisms behind host cell invasion and the development of anti-infective therapeutic strategies.
Collapse
Affiliation(s)
- Allegra Via
- Department of Physics, Sapienza University, 00185 Rome, Italy
| | - Bora Uyar
- Structural and Computational Biology, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Christine Brun
- Inserm, UMR1090 TAGC, Marseille F-13288, France; Aix-Marseille Université, UMR1090 TAGC, Marseille F-13288, France; CNRS, Marseille F-13402, France
| | - Andreas Zanzoni
- Inserm, UMR1090 TAGC, Marseille F-13288, France; Aix-Marseille Université, UMR1090 TAGC, Marseille F-13288, France.
| |
Collapse
|
34
|
Formela M, Samardakiewicz S, Marczak Ł, Nowak W, Narożna D, Bednarski W, Kasprowicz-Maluśki A, Morkunas I. Effects of endogenous signals and Fusarium oxysporum on the mechanism regulating genistein synthesis and accumulation in yellow lupine and their impact on plant cell cytoskeleton. Molecules 2014; 19:13392-421. [PMID: 25178062 PMCID: PMC6271453 DOI: 10.3390/molecules190913392] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 08/07/2014] [Accepted: 08/18/2014] [Indexed: 01/01/2023] Open
Abstract
The aim of the study was to examine cross-talk interactions of soluble sugars (sucrose, glucose and fructose) and infection caused by Fusarium oxysporum f.sp. lupini on the synthesis of genistein in embryo axes of Lupinus luteus L.cv. Juno. Genistein is a free aglycone, highly reactive and with the potential to inhibit fungal infection and development of plant diseases. As signal molecules, sugars strongly stimulated accumulation of isoflavones, including genistein, and the expression of the isoflavonoid biosynthetic genes. Infection significantly enhanced the synthesis of genistein and other isoflavone aglycones in cells of embryo axes of yellow lupine with high endogenous sugar levels. The activity of β-glucosidase, the enzyme that releases free aglycones from their glucoside bindings, was higher in the infected tissues than in the control ones. At the same time, a very strong generation of the superoxide anion radical was observed in tissues with high sugar contents already in the initial stage of infection. During later stages after inoculation, a strong generation of semiquinone radicals was observed, which level was relatively higher in tissues deficient in sugars than in those with high sugar levels. Observations of actin and tubulin cytoskeletons in cells of infected embryo axes cultured on the medium with sucrose, as well as the medium without sugar, showed significant differences in their organization.
Collapse
Affiliation(s)
- Magda Formela
- Department of Plant Physiology, Poznań University of Life Sciences, Wołyńska 35, Poznań 60-637, Poland.
| | - Sławomir Samardakiewicz
- Laboratory of Electron and Confocal Microscopy, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, Poznań 60-614, Poland.
| | - Łukasz Marczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Z. Noskowskiego 12/14, Poznań 61-704, Poland.
| | - Witold Nowak
- Laboratory of Molecular Biology Techniques, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, Poznań 60-614, Poland.
| | - Dorota Narożna
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11, Poznań 60-632, Poland.
| | - Waldemar Bednarski
- Institute of Molecular Physics, Polish Academy of Sciences, Smoluchowskiego 17, Poznań 60-179, Poland.
| | - Anna Kasprowicz-Maluśki
- Department of Molecular and Cellular Biology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, Poznań 60-614, Poland.
| | - Iwona Morkunas
- Department of Plant Physiology, Poznań University of Life Sciences, Wołyńska 35, Poznań 60-637, Poland.
| |
Collapse
|
35
|
Mittal R, Grati M, Gerring R, Blackwelder P, Yan D, Li JD, Liu XZ. In vitro interaction of Pseudomonas aeruginosa with human middle ear epithelial cells. PLoS One 2014; 9:e91885. [PMID: 24632826 PMCID: PMC3954863 DOI: 10.1371/journal.pone.0091885] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 02/14/2014] [Indexed: 01/27/2023] Open
Abstract
Background Otitis media (OM) is an inflammation of the middle ear which can be acute or chronic. Acute OM is caused by Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis whereas Pseudomonas aeruginosa is a leading cause of chronic suppurative otitis media (CSOM). CSOM is a chronic inflammatory disorder of the middle ear characterized by infection and discharge. The survivors often suffer from hearing loss and neurological sequelae. However, no information is available regarding the interaction of P. aeruginosa with human middle ear epithelial cells (HMEECs). Methodology and Findings In the present investigation, we demonstrate that P. aeruginosa is able to enter and survive inside HMEECs via an uptake mechanism that is dependent on microtubule and actin microfilaments. The actin microfilament disrupting agent as well as microtubule inhibitors exhibited significant decrease in invasion of HMEECs by P. aeruginosa. Confocal microscopy demonstrated F-actin condensation associated with bacterial entry. This recruitment of F-actin was transient and returned to normal distribution after bacterial internalization. Scanning electron microscopy demonstrated the presence of bacteria on the surface of HMEECs, and transmission electron microscopy confirmed the internalization of P. aeruginosa located in the plasma membrane-bound vacuoles. We observed a significant decrease in cell invasion of OprF mutant compared to the wild-type strain. P. aeruginosa induced cytotoxicity, as demonstrated by the determination of lactate dehydrogenase levels in culture supernatants of infected HMEECs and by a fluorescent dye-based assay. Interestingly, OprF mutant showed little cell damage compared to wild-type P. aeruginosa. Conclusions and Significance This study deciphered the key events in the interaction of P. aeruginosa with HMEECs in vitro and highlighted the role of bacterial outer membrane protein, OprF, in this process. Understanding the molecular mechanisms in the pathogenesis of CSOM will help in identifying novel targets to design effective therapeutic strategies and to prevent hearing loss.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - M’hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Robert Gerring
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Patricia Blackwelder
- Center for Advanced Microscopy, University of Miami, Coral Gables, Florida, United States of America
- RSMAS, University of Miami, Key Biscayne, Florida, United States of America
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jian-Dong Li
- Center for Inflammation, Immunity, and Infection and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
36
|
Jayamani E, Mylonakis E. Effector triggered manipulation of host immune response elicited by different pathotypes of Escherichia coli. Virulence 2014; 5:733-9. [PMID: 25513774 PMCID: PMC4189879 DOI: 10.4161/viru.29948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/08/2014] [Accepted: 07/15/2014] [Indexed: 12/28/2022] Open
Abstract
Effectors are virulence factors that are secreted by bacteria during an infection in order to subvert cellular processes or induce the surveillance system of the host. Pathogenic microorganisms encode effectors, toxins and components of secretion systems that inject the effectors to the host. Escherichia coli is part of the innocuous commensal microbial flora of the gastrointestinal tract. However, pathogenic E. coli can cause diarrheal and extraintestinal diseases. Pathogenic E. coli uses secretion systems to inject an array of effector proteins directly into the host cells. Herein, we discuss the effectors secreted by different pathotypes of E. coli and provide an overview of strategies employed by effectors to target the host cellular and subcellular processes as well as their role in triggering host immune response.
Collapse
Affiliation(s)
- Elamparithi Jayamani
- Division of Infectious Diseases; Rhode Island Hospital; Alpert Medical School of Brown University; Providence, RI USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases; Rhode Island Hospital; Alpert Medical School of Brown University; Providence, RI USA
| |
Collapse
|