1
|
El-Shiekh RA, Elshimy R. Therapeutic effects of Stemmoside C against Salmonella enterica serotype typhimurium infected BALB/c mice. Steroids 2023; 199:109296. [PMID: 37591445 DOI: 10.1016/j.steroids.2023.109296] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Salmonella is a Gram-negative bacterium that causes gastrointestinal diseases in 20 to 40 million people globally. Stemmoside C is a steroidal glycoside isolated from Argel, although its antibacterial and antibiofilm properties have not been studied. The antibacterial activity of Stemmoside C against Salmonella enterica was revealed, where MIC of the compound was 16 μg/mL (0.15 µM). Biofilm-associated Stemmoside C treatment destroyed S. typhi cells and reduced viable S. typhi numbers below detectable levels. When compared to Stemmoside C or Ciprofluxacin-treated mice, infected BALB/c mice had a greater death rate and a larger bacterial blood burden. The protective effects of orally administered Stemmoside C at dose of 25 and 50 mg/kg b.wt. against bacterial infection was associated with reduction in the levels of inflammatory cytokines (IFN-γ, Il-1β, IL-2, IL-6, MPO, and TNF-α) and elevation of anti-inflammatory cytokine (IL-10 and IL-12) in serum. Where, Stemmoside C at dose of 50 mg/kg b.wt. regulated the levels almost as normal control group and demonstrated apparently normal intestinal sections. It also resulted in a decrease in the number of viable S. typhi retrieved from feces. Stemmoside C is a promising drug for the treatment or prevention of S. typhimurium infection.
Collapse
Affiliation(s)
- Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr el Aini st., Cairo 11562, Egypt.
| | - Rana Elshimy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt; Department of Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt.
| |
Collapse
|
2
|
Chaukimath P, Frankel G, Visweswariah SS. The metabolic impact of bacterial infection in the gut. FEBS J 2023; 290:3928-3945. [PMID: 35731686 DOI: 10.1111/febs.16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/02/2022] [Accepted: 06/21/2022] [Indexed: 08/17/2023]
Abstract
Bacterial infections of the gut are one of the major causes of morbidity and mortality worldwide. The interplay between the pathogen and the host is finely balanced, with the bacteria evolving to proliferate and establish infection. In contrast, the host mounts a response to first restrict and then eliminate the infection. The intestine is a rapidly proliferating tissue, and metabolism is tuned to cater to the demands of proliferation and differentiation along the crypt-villus axis (CVA) in the gut. As bacterial pathogens encounter the intestinal epithelium, they elicit changes in the host cell, and core metabolic pathways such as the tricarboxylic acid (TCA) cycle, lipid metabolism and glycolysis are affected. This review highlights the mechanisms utilized by diverse gut bacterial pathogens to subvert host metabolism and describes host responses to the infection.
Collapse
Affiliation(s)
- Pooja Chaukimath
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Gad Frankel
- Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College, London, UK
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
3
|
Guo WH, Zhang K, Yang LH. Potential Mechanisms of Pyrrosiae Folium in Treating Prostate Cancer Based on Network Pharmacology and Molecular Docking. Drug Dev Ind Pharm 2022; 48:189-197. [PMID: 35730236 DOI: 10.1080/03639045.2022.2088785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective The network pharmacology approach and molecular docking were employed to explore the mechanism of Pyrrosiae Folium(PF) against prostate cancer (PCa). Methods The active compounds and their corresponding putative targets of PF were identified by the Traditional Chinese Medicine Systems Pharmacology (TCMSP), the gene names of the targets were obtained from the UniProt database. The collection of genes associated with PCa were obtained from GeneCards and DisGeNET database. We merged the drug targets and disease targets by online software, Draw Venn Diagram. The resulting gene list was imported into R software (v3.6.3) for GO and KEGG function enrichment analysis. The STRING database was utilized for protein-protein interaction (PPI) network construction. The cytoHubba plugin of Cytoscape was used to identify core genes. Further, molecular docking analysis of the hub targets were carried out using AutoDock Vina software (v1.5.6). Results A total of 6 active components were screened by PF, with 167 corresponding putative targets, 1395 related targets for PCa, and 113 targets for drugs and diseases. The "drug-component-disease-target" network was constructed by Cytoscape software and the target genes mainly involved in the complex treating effects associated with response to oxidative stress, cytokine activity, pathways in cancer, prostate cancer pathway and TNF signaling pathway. Core genes in the PPI network were TNF, JUN, IL6, IL1B, CXCL8, RELA, CCL2, TP53, IL10 and FOS. The molecular docking results reveal the better binding affinity of 6 active components to the core targets. Conclusion The results of this study indicated that PF may be have a certain anti-PCa effect by regulating related target genes, affecting Pathways in cancer, TNF signaling pathway, Hepatitis B signaling pathway.
Collapse
Affiliation(s)
- Wen-Hua Guo
- Modern College of Humanities and Science of Shanxi Normal University, Linfen, Shanxi 041004, P.R. China.,School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Kun Zhang
- School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Lu-Hong Yang
- Modern College of Humanities and Science of Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| |
Collapse
|
4
|
Genetic Polymorphisms of IGF1 and IGF1R Genes and Their Effects on Growth Traits in Hulun Buir Sheep. Genes (Basel) 2022; 13:genes13040666. [PMID: 35456472 PMCID: PMC9031115 DOI: 10.3390/genes13040666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
The identification of candidate genes and genetic variations associated with growth traits is important for sheep breeding. Insulin like growth factor 1 (IGF1) and insulin like growth factor 1 receptor (IGF1R) are well-accepted candidate genes that affect animal growth and development. The current study attempted to assess the association between IGF1 and IGF1R genetic polymorphisms and growth traits in Hulun Buir sheep. To achieve this goal, we first identified three and ten single nucleotide polymorphisms (SNPs) in exons of IGF1 and IGF1R in Hulun Buir sheep and then constructed six haplotypes of IGF1R based on linkage disequilibrium, respectively. Association studies were performed between SNPs and haplotypes of IGF1 and IGF1R with twelve growth traits in a population encompassing 229 Hulun Buir sheep using a general linear model. Our result indicated three SNPs in IGF1 were significantly associated with four growth traits (p < 0.05). In IGF1R, three SNPs and two haplotype blocks were significantly associated with twelve growth traits (p < 0.05). The combined haplotype H5H5 and H5H6 in IGF1R showed the strong association with 12 superior growth traits in Hulun Buir sheep (p < 0.05). In conclusion, we identified SNPs and haplotype combinations associated with the growth traits, which provided genetic resources for marker-assisted selection (MAS) in Hulun Buir sheep breeding.
Collapse
|
5
|
Identification of Somatostatin Receptor Subtype 1 (SSTR1) Gene Polymorphism and Their Association with Growth Traits in Hulun Buir Sheep. Genes (Basel) 2021; 13:genes13010077. [PMID: 35052417 PMCID: PMC8775034 DOI: 10.3390/genes13010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
This study was conducted to evaluate SSTR1 gene polymorphisms and their association with growth traits in Hulun Buir sheep. We followed 233 Hulun Buir sheep from birth to 16 months of age, born in the same pasture and on the same year under a consistent grazing conditions. The body weight (BW), body height (BH), body length (BL), chest circumference (ChC), chest depth (ChD), chest width (ChW), hip width (HW), and cannon circumference (CaC) were measured and recorded at birth, 4 months, 9 months, and 16 months of age. The polymorphisms of the SSTR1 gene in Hulun Buir sheep were excavated using exon sequencing, and association analyses of between SNPs and growth traits at each growth stage were conducted. The results showed that there were four SNPs in Exon 2 of the SSTR1 gene, SNP1, SNP2, and SNP3 were low mutation sites, and SNP4 was a moderate mutation site. Four SNPs were consistent with Hardy–Weinberg equilibrium, and all of them were synonymous mutations. The association analyses found that the genotypes of SNP2 were significantly associated with WW and BH at 4 months of age, BW, BL, ChC, and HW at 9 months of age (p < 0.05), and extremely significantly associated with ChD at 4 and 9 months of age (p < 0.01). There were significant associations between SNP3 and BH at 9 months of age, between SNP4 and ChD, ChW, and CaC at 9 months of age, and BW and ChC at 16 months of age (p < 0.05). There were no detectable associations with growth traits among the seven haplotypes between the SNP1, 3, and 4 of a strong linkage disequilibrium (p > 0.05). These results indicated that SNP2, SNP3, and SNP4 may be used as molecular markers for growth traits of Hulun Buir sheep.
Collapse
|
6
|
A Bioinformatics Approach to Identifying Potential Biomarkers for Cryptosporidium parvum: A Coccidian Parasite Associated with Fetal Diarrhea. Vaccines (Basel) 2021; 9:vaccines9121427. [PMID: 34960172 PMCID: PMC8705633 DOI: 10.3390/vaccines9121427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 01/07/2023] Open
Abstract
Cryptosporidium parvum (C. parvum) is a protozoan parasite known for cryptosporidiosis in pre-weaned calves. Animals and patients with immunosuppression are at risk of developing the disease, which can cause potentially fatal diarrhoea. The present study aimed to construct a network biology framework based on the differentially expressed genes (DEGs) of C. parvum infected subjects. In this way, the gene expression profiling analysis of C. parvum infected individuals can give us a snapshot of actively expressed genes and transcripts under infection conditions. In the present study, we have analyzed microarray data sets and compared the gene expression profiles of the patients with the different data sets of the healthy control. Using a network medicine approach to identify the most influential genes in the gene interaction network, we uncovered essential genes and pathways related to C. parvum infection. We identified 164 differentially expressed genes (109 up- and 54 down-regulated DEGs) and allocated them to pathway and gene set enrichment analysis. The results underpin the identification of seven significant hub genes with high centrality values: ISG15, MX1, IFI44L, STAT1, IFIT1, OAS1, IFIT3, RSAD2, IFITM1, and IFI44. These genes are associated with diverse biological processes not limited to host interaction, type 1 interferon production, or response to IL-gamma. Furthermore, four genes (IFI44, IFIT3, IFITM1, and MX1) were also discovered to be involved in innate immunity, inflammation, apoptosis, phosphorylation, cell proliferation, and cell signaling. In conclusion, these results reinforce the development and implementation of tools based on gene profiles to identify and treat Cryptosporidium parvum-related diseases at an early stage.
Collapse
|
7
|
Salmonella Typhimurium impairs glycolysis-mediated acidification of phagosomes to evade macrophage defense. PLoS Pathog 2021; 17:e1009943. [PMID: 34555129 PMCID: PMC8491875 DOI: 10.1371/journal.ppat.1009943] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/05/2021] [Accepted: 09/07/2021] [Indexed: 11/19/2022] Open
Abstract
Regulation of cellular metabolism is now recognized as a crucial mechanism for the activation of innate and adaptive immune cells upon diverse extracellular stimuli. Macrophages, for instance, increase glycolysis upon stimulation with pathogen-associated molecular patterns (PAMPs). Conceivably, pathogens also counteract these metabolic changes for their own survival in the host. Despite this dynamic interplay in host-pathogen interactions, the role of immunometabolism in the context of intracellular bacterial infections is still unclear. Here, employing unbiased metabolomic and transcriptomic approaches, we investigated the role of metabolic adaptations of macrophages upon Salmonella enterica serovar Typhimurium (S. Typhimurium) infections. Importantly, our results suggest that S. Typhimurium abrogates glycolysis and its modulators such as insulin-signaling to impair macrophage defense. Mechanistically, glycolysis facilitates glycolytic enzyme aldolase A mediated v-ATPase assembly and the acidification of phagosomes which is critical for lysosomal degradation. Thus, impairment in the glycolytic machinery eventually leads to decreased bacterial clearance and antigen presentation in murine macrophages (BMDM). Collectively, our results highlight a vital molecular link between metabolic adaptation and phagosome maturation in macrophages, which is targeted by S. Typhimurium to evade cell-autonomous defense.
Collapse
|
8
|
Wang Y, Wu C, Gao J, Du X, Chen X, Zhang M. Host metabolic shift during systemic Salmonella infection revealed by comparative proteomics. Emerg Microbes Infect 2021; 10:1849-1861. [PMID: 34461813 PMCID: PMC8451668 DOI: 10.1080/22221751.2021.1974316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a food-borne bacterium that causes acute gastroenteritis in humans and typhoid fever in mice. Salmonella pathogenicity island II (SPI-2) is an important virulence gene cluster responsible for Salmonella survival and replication within host cells, leading to systemic infection. Previous studies have suggested that SPI-2 function to modulate host vesicle trafficking and immune response to promote systemic infection. However, the molecular mechanism and the host responses triggered by SPI-2 remain largely unknown. To assess the roles of SPI-2, we used a differential proteomic approach to analyse host proteins levels during systemic infections in mice. Our results showed that infection by WT S. Typhimurium triggered the reprogramming of host cell metabolism and inflammatory response. Salmonella systemic infection induces an up-regulation of glycolytic process and a repression of the tricarboxylic acid (TCA) cycle. WT-infected tissues prefer to produce adenosine 5′-triphosphate (ATP) through aerobic glycolysis rather than relying on oxidative phosphorylation to generate energy. Moreover, our data also revealed that infected macrophages may undergo both M1 and M2 polarization. In addition, our results further suggest that SPI-2 is involved in altering actin cytoskeleton to facilitate the Salmonella-containing vacuole (SCV) biogenesis and perhaps even the release of bacteria later in the infection process. Results from our study provide valuable insights into the roles of SPI-2 during systemic Salmonella infection and will guide future studies to dissect the molecular mechanisms of how SPI-2 functions in vivo.
Collapse
Affiliation(s)
- Yuanyuan Wang
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Chunmei Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Jiacong Gao
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Xudong Du
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Xiangyun Chen
- TEDA School of Biological Sciences and Biotechnology, Nankai University, Tianjin, People's Republic of China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| |
Collapse
|
9
|
Jiang L, Wang P, Song X, Zhang H, Ma S, Wang J, Li W, Lv R, Liu X, Ma S, Yan J, Zhou H, Huang D, Cheng Z, Yang C, Feng L, Wang L. Salmonella Typhimurium reprograms macrophage metabolism via T3SS effector SopE2 to promote intracellular replication and virulence. Nat Commun 2021; 12:879. [PMID: 33563986 PMCID: PMC7873081 DOI: 10.1038/s41467-021-21186-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Salmonella Typhimurium establishes systemic infection by replicating in host macrophages. Here we show that macrophages infected with S. Typhimurium exhibit upregulated glycolysis and decreased serine synthesis, leading to accumulation of glycolytic intermediates. The effects on serine synthesis are mediated by bacterial protein SopE2, a type III secretion system (T3SS) effector encoded in pathogenicity island SPI-1. The changes in host metabolism promote intracellular replication of S. Typhimurium via two mechanisms: decreased glucose levels lead to upregulated bacterial uptake of 2- and 3-phosphoglycerate and phosphoenolpyruvate (carbon sources), while increased pyruvate and lactate levels induce upregulation of another pathogenicity island, SPI-2, known to encode virulence factors. Pharmacological or genetic inhibition of host glycolysis, activation of host serine synthesis, or deletion of either the bacterial transport or signal sensor systems for those host glycolytic intermediates impairs S. Typhimurium replication or virulence. Salmonella Typhimurium establishes systemic infection by replicating in host macrophages. Here, Jiang et al. show that infected macrophages exhibit upregulated glycolysis and decreased serine synthesis, leading to accumulation of glycolytic intermediates that promote intracellular replication and virulence of S. Typhimurium.
Collapse
Affiliation(s)
- Lingyan Jiang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Peisheng Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xiaorui Song
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Huan Zhang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Shuangshuang Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Jingting Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Wanwu Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Runxia Lv
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xiaoqian Liu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Shuai Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Jiaqi Yan
- College of Life Sciences, Nankai University, Tianjin, China
| | - Haiyan Zhou
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Di Huang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Zhihui Cheng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Chen Yang
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Lu Feng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China. .,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China.
| | - Lei Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China. .,TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China. .,The Institute of Translational Medicine Research, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Nankai University, Tianjin, China.
| |
Collapse
|
10
|
Miyazawa K, Nakai D, Nakamura Y, Tatsuno T, Inoue S, Nakazawa Y, Ishigaki Y. Effects of the xanthine oxidase inhibitor, febuxostat, on the expression of monocyte chemoattractant protein-1 and synchronous genes in MDCK cells treated with calcium oxalate monohydrate crystals. Int J Urol 2021; 28:339-345. [PMID: 33393162 DOI: 10.1111/iju.14450] [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: 08/19/2020] [Accepted: 11/03/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To examine the effects of the selective xanthine oxidase inhibitor febuxostat on the expression of inflammation-related genes involved in stone formation. METHODS Madin-Darby canine kidney cells were exposed to febuxostat, followed by calcium oxalate monohydrate crystals. Monocyte chemoattractant protein-1 messenger ribonucleic acid expression levels were determined by real-time reverse transcription polymerase chain reaction analysis. Deoxyribonucleic acid microarray analysis was utilized to evaluate gene expression. RESULTS Calcium oxalate monohydrate crystals activated monocyte chemoattractant protein-1 messenger ribonucleic acid expression in a time- and concentration-dependent manner. Febuxostat suppressed monocyte chemoattractant protein-1 expression. The expression levels of a group of inflammatory genes, including interleukin-8 and chemokine (C-X-C motif) ligand 10, which are downstream of reactive oxygen species, fluctuated similarly to the observed monocyte chemoattractant protein-1 fluctuations and were reduced by febuxostat pretreatment. CONCLUSIONS Febuxostat exerts preventive effects against reactive oxygen species production and oxidative stress, and might represent a potential treatment for calcium oxalate stones. In the present study, febuxostat downregulated the calcium oxalate monohydrate crystal-induced monocyte chemoattractant protein-1 messenger ribonucleic acid expression.
Collapse
Affiliation(s)
- Katsuhito Miyazawa
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Dan Nakai
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Takanori Tatsuno
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Shinya Inoue
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yusuke Nakazawa
- Department of Urology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
11
|
Wilson KM, Rodrigues DR, Briggs WN, Duff AF, Chasser KM, Bottje WG, Bielke LR. Impact of in ovo administered pioneer colonizers on intestinal proteome on day of hatch. Poult Sci 2020; 99:1254-1266. [PMID: 32111303 PMCID: PMC7587751 DOI: 10.1016/j.psj.2019.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pioneer colonization of the gastrointestinal tract (GIT) by bacteria is thought to have major influence on neonatal tissue development. Previous studies have shown in ovo inoculation of embryos with saline (S), species of Citrobacter (C, C2), or lactic acid bacteria (L) resulted in an altered microbiome on day of the hatch (DOH). The present study investigated GIT proteomic changes at DOH in relation to different inoculations. Embryos were inoculated in ovo with S or ∼102 cfu of C, C2, or L at 18 embryonic days. On DOH, the GIT was collected, and tissue proteins were extracted for analysis via tandem mass spectrometry. A total of 493 proteins were identified for differential comparison with S at P ≤ 0.10. Different levels were noted in 107, 39, and 78 proteins in C, C2, and L groups, respectively, which were uploaded to Ingenuity Pathway Analysis to determine canonical pathways and biological functions related to these changes. Three members of the cytokine family (interleukin [IL]-1β, IL6, and Oncostatin M) were predicted to be activated in C2, indicated with Z-score ≥ 1.50, which suggested an overall proinflammatory GIT condition. This was consistent with the activation of the acute-phase response signaling pathway seen exclusively in C2 (Z-score = 2.00, P < 0.01). However, activation (Z-score = 2.00) of IL-13, upregulation of peroxiredoxin-1 and superoxide dismutase 1, in addition to activation of nitric oxide signaling in the cardiovascular system of the L treatment may predict a state of increased antioxidant capacity and decreased inflammatory status. The nuclear factor erythroid 2-related factor 2 (NRF2)-mediated oxidative stress response (Z-score = 2.00, P < 0.01) was predicted to be upregulated in C which suggested that chicks were in an inflammatory state and associated oxidative stress, but the impact of these pathways differed from that of C2. These changes in the proteome suggest that pioneer colonizing microbiota may have a strong impact on pathways associated with GIT immune and cellular development.
Collapse
Affiliation(s)
- K M Wilson
- Department of Animal Science, The Ohio State University, Columbus, OH
| | - D R Rodrigues
- Department of Animal Science, The Ohio State University, Columbus, OH
| | - W N Briggs
- Department of Animal Science, The Ohio State University, Columbus, OH
| | - A F Duff
- Department of Animal Science, The Ohio State University, Columbus, OH
| | - K M Chasser
- Department of Animal Science, The Ohio State University, Columbus, OH
| | - W G Bottje
- Department of Poultry Science, University of Arkansas, Fayetteville, AR
| | - L R Bielke
- Department of Animal Science, The Ohio State University, Columbus, OH.
| |
Collapse
|
12
|
Benis N, Wells JM, Smits MA, Kar SK, van der Hee B, Dos Santos VAPM, Suarez-Diez M, Schokker D. High-level integration of murine intestinal transcriptomics data highlights the importance of the complement system in mucosal homeostasis. BMC Genomics 2019; 20:1028. [PMID: 31888466 PMCID: PMC6937694 DOI: 10.1186/s12864-019-6390-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 12/12/2019] [Indexed: 12/25/2022] Open
Abstract
Background The mammalian intestine is a complex biological system that exhibits functional plasticity in its response to diverse stimuli to maintain homeostasis. To improve our understanding of this plasticity, we performed a high-level data integration of 14 whole-genome transcriptomics datasets from samples of intestinal mouse mucosa. We used the tool Centrality based Pathway Analysis (CePa), along with information from the Reactome database. Results The results show an integrated response of the mouse intestinal mucosa to challenges with agents introduced orally that were expected to perturb homeostasis. We observed that a common set of pathways respond to different stimuli, of which the most reactive was the Regulation of Complement Cascade pathway. Altered expression of the Regulation of Complement Cascade pathway was verified in mouse organoids challenged with different stimuli in vitro. Conclusions Results of the integrated transcriptomics analysis and data driven experiment suggest an important role of epithelial production of complement and host complement defence factors in the maintenance of homeostasis.
Collapse
Affiliation(s)
- Nirupama Benis
- Host Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands. .,Systems and Synthetic Biology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Jerry M Wells
- Host Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands
| | - Mari A Smits
- Host Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands.,Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands.,Wageningen Bioveterinary Research, Wageningen University, Wageningen, The Netherlands
| | - Soumya Kanti Kar
- Host Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands.,Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Bart van der Hee
- Host Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands
| | - Vitor A P Martins Dos Santos
- Systems and Synthetic Biology, Wageningen University & Research, Wageningen, The Netherlands.,LifeGlimmer GmbH, Berlin, Germany
| | - Maria Suarez-Diez
- Systems and Synthetic Biology, Wageningen University & Research, Wageningen, The Netherlands
| | - Dirkjan Schokker
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
13
|
Sang D, Bai S, Yin S, Jiang S, Ye L, Hou W, Yao Y, Wang H, Shen Y, Shen B, Du J. Role of TRPP2 in mouse airway smooth muscle tension and respiration. Am J Physiol Lung Cell Mol Physiol 2019; 317:L466-L474. [PMID: 31411061 DOI: 10.1152/ajplung.00513.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The transient receptor potential polycystin-2 (TRPP2) is encoded by the Pkd2 gene, and mutation of this gene can cause autosomal dominant polycystic kidney disease (ADPKD). Some patients with ADPKD experience extrarenal manifestations, including radiologic and clinical bronchiectasis. We hypothesized that TRPP2 may regulate airway smooth muscle (ASM) tension. Thus, we used smooth muscle-Pkd2 conditional knockout (Pkd2SM-CKO) mice to investigate whether TRPP2 regulated ASM tension and whether TRPP2 deficiency contributed to bronchiectasis associated with ADPKD. Compared with wild-type mice, Pkd2SM-CKO mice breathed more shallowly and faster, and their cross-sectional area ratio of bronchi to accompanying pulmonary arteries was higher, suggesting that TRPP2 may regulate ASM tension and contribute to the occurrence of bronchiectasis in ADPKD. In a bioassay examining isolated tracheal ring tension, no significant difference was found for high-potassium-induced depolarization of the ASM between the two groups, indicating that TRPP2 does not regulate depolarization-induced ASM contraction. By contrast, carbachol-induced contraction of the ASM derived from Pkd2SM-CKO mice was significantly reduced compared with that in wild-type mice. In addition, relaxation of the carbachol-precontracted ASM by isoprenaline, a β-adrenergic receptor agonist that acts through the cAMP/adenylyl cyclase pathway, was also significantly attenuated in Pkd2SM-CKO mice compared with that in wild-type mice. Thus, TRPP2 deficiency suppressed both contraction and relaxation of the ASM. These results provide a potential target for regulating ASM tension and for developing therapeutic alternatives for some ADPKD complications of the respiratory system or for independent respiratory disease, especially bronchiectasis.
Collapse
Affiliation(s)
- Dacheng Sang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Orthopedic Surgery, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Suwen Bai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Sheng Yin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Neurosurgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Sen Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Neurosurgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Li Ye
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wenxuan Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yanheng Yao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haoran Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yonggang Shen
- Nursing Faculty, Anhui Health College, Chizhou, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
14
|
Escoll P, Buchrieser C. Metabolic reprogramming: an innate cellular defence mechanism against intracellular bacteria? Curr Opin Immunol 2019; 60:117-123. [PMID: 31247377 DOI: 10.1016/j.coi.2019.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
The limited metabolic resources of a cell represent an intriguing 'conflict of interest' during host-pathogen interactions, as the battle for nutrients might determine the outcome of an infection. To adapt their metabolic needs, innate immune cells such as monocytes, macrophages or dendritic cells reprogram their metabolism upon activation by microbial compounds. In turn, infection by intracellular bacteria provokes metabolic alterations of the host cell that benefit the pathogen. Here, we discuss the state-of-the-art knowledge on metabolic reprogramming of host cells upon activation or infection with intracellular bacteria. The study of the host-driven and pathogen-driven metabolic alterations that seem to co-exist during infection is an emerging field that will define the metabolic pathways that might be targeted to combat infection.
Collapse
Affiliation(s)
- Pedro Escoll
- Institut Pasteur, Biologie des Bactéries Intracellulaires, UMR 3525, CNRS, Paris, France.
| | - Carmen Buchrieser
- Institut Pasteur, Biologie des Bactéries Intracellulaires, UMR 3525, CNRS, Paris, France.
| |
Collapse
|
15
|
Sui G, Cheng G, Yuan J, Hou X, Kong X, Niu H. Interleukin (IL)-13, Prostaglandin E2 (PGE2), and Prostacyclin 2 (PGI2) Activate Hepatic Stellate Cells via Protein kinase C (PKC) Pathway in Hepatic Fibrosis. Med Sci Monit 2018; 24:2134-2141. [PMID: 29633755 PMCID: PMC5909417 DOI: 10.12659/msm.906442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Protein kinase C (PKC), interleukin (IL)-13, prostaglandin E2 (PGE2), and prostacyclin 2 (PGI2) can all play crucial roles in pulmonary fibrosis. However, their functions remain unclear in hepatic fibrosis mediated by hepatic stellate cells (HSCs), which has been demonstrated to be related to transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF). MATERIAL AND METHODS All the experiments were based on LX-2 Hepatic stellate cells. The expression of TGF-β1 and PDGF were assessed by ELISA, RT-PCR, and Western blotting in human HSCs treated by IL-13, PGE2, and PGI2, respectively. At the same time, bridge assay and CCK8 assay were used to detect the cell proliferation and activity, PKC activity assay was used to test the activity of PKC, and PKC agonist and antagonist were used to verify the results obtained previously. RESULTS We found that IL-13, PGE2, and PGI2 significantly enhanced the expression of TGF-β1 and PDGF in human HSCs, which also clearly improved the proliferation and cell activity of HSCs. Moreover, PKC activity was significantly increased following IL-13, PGE2, and PGI2 treatments. We also found that the expression of TGF-β1 and PDGF, as well as the proliferation and cell activity of HSCs, were significantly enhanced by the PKC agonist phorbol 12-myristate 13-acetate (PMA), but suppressed by the PKC antagonist calphostin C. CONCLUSIONS We found that IL-13, PGE2, and PGI2 stimulated HSCs proliferation and secretion of TGF-β1 and PDGF by activating PKC, which predicted their potential roles in hepatic fibrosis.
Collapse
Affiliation(s)
- Guode Sui
- Department of Emergency General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Guang Cheng
- Department of Emergency General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Junjun Yuan
- Department of Emergency General Surgery, Affiliated Hospital of Qingdao University, Shandong, China (mainland)
| | - Xuena Hou
- Department of Emergency General Surgery, Affiliated Hospital of Qingdao University, Shandong, China (mainland)
| | - Xiaochen Kong
- Department of Emergency General Surgery, Affiliated Hospital of Qingdao University, Shandong, China (mainland)
| | - Haitao Niu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
16
|
Cabezas-Cruz A, Espinosa PJ, Obregón DA, Alberdi P, de la Fuente J. Ixodes scapularis Tick Cells Control Anaplasma phagocytophilum Infection by Increasing the Synthesis of Phosphoenolpyruvate from Tyrosine. Front Cell Infect Microbiol 2017; 7:375. [PMID: 28861402 PMCID: PMC5562928 DOI: 10.3389/fcimb.2017.00375] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/04/2017] [Indexed: 01/14/2023] Open
Abstract
The obligate intracellular pathogen, Anaplasma phagocytophilum, is the causative agent of life-threatening diseases in humans and animals. A. phagocytophilum is an emerging tick-borne pathogen in the United States, Europe, Africa and Asia, with increasing numbers of infected people and animals every year. It is increasingly recognized that intracellular pathogens modify host cell metabolic pathways to increase infection and transmission in both vertebrate and invertebrate hosts. Recent reports have shown that amino acids are central to the host–pathogen metabolic interaction. In this study, a genome-wide search for components of amino acid metabolic pathways was performed in Ixodes scapularis, the main tick vector of A. phagocytophilum in the United States, for which the genome was recently published. The enzymes involved in the synthesis and degradation pathways of the twenty amino acids were identified. Then, the available transcriptomics and proteomics data was used to characterize the mRNA and protein levels of I. scapularis amino acid metabolic pathway components in response to A. phagocytophilum infection of tick tissues and ISE6 tick cells. Our analysis was focused on the interplay between carbohydrate and amino acid metabolism during A. phagocytophilum infection in ISE6 cells. The results showed that tick cells increase the synthesis of phosphoenolpyruvate (PEP) from tyrosine to control A. phagocytophilum infection. Metabolic pathway analysis suggested that this is achieved by (i) increasing the transcript and protein levels of mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M), (ii) shunting tyrosine into the tricarboxylic acid (TCA) cycle to increase fumarate and oxaloacetate which will be converted into PEP by PEPCK-M, and (iii) blocking all the pathways that use PEP downstream gluconeogenesis (i.e., de novo serine synthesis pathway (SSP), glyceroneogenesis and gluconeogenesis). While sequestering host PEP may be critical for this bacterium because it cannot actively carry out glycolysis to produce PEP, excess of this metabolite may be toxic for A. phagocytophilum. The present work provides a more comprehensive view of the major amino acid metabolic pathways involved in the response to pathogen infection in ticks, and provides the basis for further studies to develop novel strategies for the control of granulocytic anaplasmosis.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Biologie Moléculaire et Immunologie Parasitaires (BIPAR), Unité Mixte de Recherche (UMR), Institut National Recherche Agronomique, Agence Nationale Sécurité Sanitaire Alimentaire Nationale (ANSES), Ecole Nationale Vétérinaire d'Alfort, Université Paris-EstMaisons-Alfort, France.,Department of Parasitology, Faculty of Science, University of South BohemiaČeské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of SciencesČeské Budějovice, Czechia
| | - Pedro J Espinosa
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - Dasiel A Obregón
- Cell and Molecular Biology Laboratory, University of Sao PauloSao Paulo, Brazil
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM)Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, United States
| |
Collapse
|
17
|
Mass spectrometry imaging identifies palmitoylcarnitine as an immunological mediator during Salmonella Typhimurium infection. Sci Rep 2017; 7:2786. [PMID: 28584281 PMCID: PMC5459799 DOI: 10.1038/s41598-017-03100-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Salmonella Typhimurium causes a self-limiting gastroenteritis that may lead to systemic disease. Bacteria invade the small intestine, crossing the intestinal epithelium from where they are transported to the mesenteric lymph nodes (MLNs) within migrating immune cells. MLNs are an important site at which the innate and adaptive immune responses converge but their architecture and function is severely disrupted during S. Typhimurium infection. To further understand host-pathogen interactions at this site, we used mass spectrometry imaging (MSI) to analyse MLN tissue from a murine model of S. Typhimurium infection. A molecule, identified as palmitoylcarnitine (PalC), was of particular interest due to its high abundance at loci of S. Typhimurium infection and MLN disruption. High levels of PalC localised to sites within the MLNs where B and T cells were absent and where the perimeter of CD169+ sub capsular sinus macrophages was disrupted. MLN cells cultured ex vivo and treated with PalC had reduced CD4+CD25+ T cells and an increased number of B220+CD19+ B cells. The reduction in CD4+CD25+ T cells was likely due to apoptosis driven by increased caspase-3/7 activity. These data indicate that PalC significantly alters the host response in the MLNs, acting as a decisive factor in infection outcome.
Collapse
|
18
|
Kommadath A, Bao H, Choi I, Reecy JM, Koltes JE, Fritz-Waters E, Eisley CJ, Grant JR, Rowland RRR, Tuggle CK, Dekkers JCM, Lunney JK, Guan LL, Stothard P, Plastow GS. Genetic architecture of gene expression underlying variation in host response to porcine reproductive and respiratory syndrome virus infection. Sci Rep 2017; 7:46203. [PMID: 28393889 PMCID: PMC5385538 DOI: 10.1038/srep46203] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 03/13/2017] [Indexed: 01/21/2023] Open
Abstract
It has been shown that inter-individual variation in host response to porcine reproductive and respiratory syndrome (PRRS) has a heritable component, yet little is known about the underlying genetic architecture of gene expression in response to PRRS virus (PRRSV) infection. Here, we integrated genome-wide genotype, gene expression, viremia level, and weight gain data to identify genetic polymorphisms that are associated with variation in inter-individual gene expression and response to PRRSV infection in pigs. RNA-seq analysis of peripheral blood samples collected just prior to experimental challenge (day 0) and at 4, 7, 11 and 14 days post infection from 44 pigs revealed 6,430 differentially expressed genes at one or more time points post infection compared to the day 0 baseline. We mapped genetic polymorphisms that were associated with inter-individual differences in expression at each day and found evidence of cis-acting expression quantitative trait loci (cis-eQTL) for 869 expressed genes (qval < 0.05). Associations between cis-eQTL markers and host response phenotypes using 383 pigs suggest that host genotype-dependent differences in expression of GBP5, GBP6, CCHCR1 and CMPK2 affect viremia levels or weight gain in response to PRRSV infection.
Collapse
Affiliation(s)
- Arun Kommadath
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
| | - Hua Bao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
- Department of Research and Development, Geneseeq Technology Inc., Toronto M5G 1L7, ON, Canada
| | - Igseo Choi
- USDA-ARS, BARC, APDL, Building1040, Beltsville 20705, MD, USA
| | - James M. Reecy
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
| | - James E. Koltes
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
- Department of Animal Science, University of Arkansas, AFLS B106D, Fayetteville, AR, 72703, USA
| | - Elyn Fritz-Waters
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
| | - Chris J. Eisley
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
- Department of Statistics, Iowa State University, 1121 Snedecor Hall, Ames, IA 50011, USA
| | - Jason R. Grant
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
| | - Robert R. R. Rowland
- College of Veterinary Medicine, Kansas State University, K-231 Mosier Hall, Manhattan 66506, KS, USA
| | - Christopher K. Tuggle
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
| | - Jack C. M. Dekkers
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, Ames 50011, IA, USA
| | - Joan K. Lunney
- USDA-ARS, BARC, APDL, Building1040, Beltsville 20705, MD, USA
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
| | - Paul Stothard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
| | - Graham S. Plastow
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, AB, Canada
| |
Collapse
|
19
|
Miarelli M, Drumo R, Signorelli F, Marchitelli C, Pavone S, Pesciaroli M, Ruggieri J, Chirullo B, Ammendola S, Battistoni A, Alborali GL, Manuali E, Pasquali P. Salmonella Typhimurium infection primes a nutriprive mechanism in piglets. Vet Microbiol 2016; 186:117-25. [DOI: 10.1016/j.vetmic.2016.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
|
20
|
Uribe JH, Collado-Romero M, Zaldívar-López S, Arce C, Bautista R, Carvajal A, Cirera S, Claros MG, Garrido JJ. Transcriptional analysis of porcine intestinal mucosa infected with Salmonella Typhimurium revealed a massive inflammatory response and disruption of bile acid absorption in ileum. Vet Res 2016; 47:11. [PMID: 26738723 PMCID: PMC4704413 DOI: 10.1186/s13567-015-0286-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/13/2015] [Indexed: 01/16/2023] Open
Abstract
Infected pork meat is an important source of non-typhoidal human salmonellosis. Understanding of molecular mechanisms involved in disease pathogenesis is important for the development of therapeutic and preventive strategies. Thus, hereby we study the transcriptional profiles along the porcine intestine during infection with Salmonella Typhimurium, as well as post-transcriptional gene modulation by microRNAs (miRNA). Sixteen piglets were orally challenged with S. Typhimurium. Samples from jejunum, ileum and colon, collected 1, 2 and 6 days post infection (dpi) were hybridized to mRNA and miRNA expression microarrays and analyzed. Jejunum showed a reduced transcriptional response indicating mild inflammation only at 2 dpi. In ileum inflammatory genes were overexpressed (e.g., IL-1B, IL-6, IL-8, IL1RAP, TNFα), indicating a strong immune response at all times of infection. Infection also down-regulated genes of the FXR pathway (e.g., NR1H4, FABP6, APOA1, SLC10A2), indicating disruption of the bile acid absorption in ileum. This result was confirmed by decreased high-density lipoprotein cholesterol in serum of infected pigs. Ileal inflammatory gene expression changes peaked at 2 dpi and tended to resolve at 6 dpi. Furthermore, miRNA analysis of ileum at 2 dpi revealed 62 miRNAs potentially regulating target genes involved in this inflammatory process (e.g., miR-374 and miR-451). In colon, genes involved in epithelial adherence, proliferation and cellular reorganization were down-regulated at 2 and 6 dpi. In summary, here we show the transcriptional changes occurring at the intestine at different time points of the infection, which are mainly related to inflammation and disruption of the bile acid metabolism.
Collapse
Affiliation(s)
- Juber Herrera Uribe
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| | - Melania Collado-Romero
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| | - Sara Zaldívar-López
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| | - Cristina Arce
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| | - Rocío Bautista
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, 29590, Málaga, Spain.
| | - Ana Carvajal
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071, León, Spain.
| | - Susanna Cirera
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Copenhagen, Denmark.
| | - M Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, 29590, Málaga, Spain.
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071, Málaga, Spain.
| | - Juan J Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, 14047, Córdoba, Spain.
| |
Collapse
|
21
|
Co-Transcriptomes of Initial Interactions In Vitro between Streptococcus Pneumoniae and Human Pleural Mesothelial Cells. PLoS One 2015; 10:e0142773. [PMID: 26566142 PMCID: PMC4643877 DOI: 10.1371/journal.pone.0142773] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/27/2015] [Indexed: 01/21/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a major causative organism of empyema, an inflammatory condition occurring in the pleural sac. In this study, we used human and Spn cDNA microarrays to characterize the transcriptional responses occurring during initial contact between Spn and a human pleural mesothelial cell line (PMC) in vitro. Using stringent filtering criteria, 42 and 23 Spn genes were up-and down-regulated respectively. In particular, genes encoding factors potentially involved in metabolic processes and Spn adherence to eukaryotic cells were up-regulated e.g. glnQ, glnA, aliA, psaB, lytB and nox. After Spn initial contact, 870 human genes were differentially regulated and the largest numbers of significant gene expression changes were found in canonical pathways for eukaryotic initiation factor 2 signaling (60 genes out of 171), oxidative phosphorylation (32/103), mitochondrial dysfunction (37/164), eIF4 and p70S6K signaling (28/142), mTOR signaling (27/182), NRF2-mediated oxidative stress response (20/177), epithelial adherens junction remodeling (11/66) and ubiquitination (22/254). The cellular response appeared to be directed towards host cell survival and defense. Spn did not activate NF-kB or phosphorylate p38 MAPK or induce cytokine production from PMC. Moreover, Spn infection of TNF-α pre-stimulated PMC inhibited production of IL-6 and IL-8 secretion by >50% (p<0.01). In summary, this descriptive study provides datasets and a platform for examining further the molecular mechanisms underlying the pathogenesis of empyema.
Collapse
|
22
|
Wu G, Liu L, Qi Y, Sun Y, Yang N, Xu G, Zhou H, Li X. Splenic gene expression profiling in White Leghorn layer inoculated with the Salmonella enterica serovar Enteritidis. Anim Genet 2015; 46:617-26. [PMID: 26358731 DOI: 10.1111/age.12341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2015] [Indexed: 01/19/2023]
Abstract
Salmonella enterica serovar Enteritidis (SE) is a foodborne pathogen that can threaten human health through contaminated poultry products. Live poultry, chicken eggs and meat are primary sources of human salmonellosis. To understand the genetic resistance of egg-type chickens in response to SE inoculation, global gene expression in the spleen of 20-week-old White Leghorn was measured using the Agilent 4 × 44 K chicken microarray at 7 and 14 days following SE inoculation (dpi). Results showed that there were 1363 genes significantly differentially expressed between inoculated and non-inoculated groups at 7 dpi (I7/N7), of which 682 were up-regulated and 681 were down-regulated genes. By contrast, 688 differentially expressed genes were observed at 14 dpi (I14/N14), of which 371 were up-regulated genes and 317 were down-regulated genes. There were 33 and 28 immune-related genes significantly differentially expressed in the comparisons of I7/N7 and I14/N14 respectively. Functional annotation revealed that several Gene Ontology (GO) terms related to immunity were significantly enriched between the inoculated and non-inoculated groups at 14 dpi but not at 7 dpi, despite a similar number of immune-related genes identified between I7/N7 and I14/N14. The immune response to SE inoculation changes with different time points following SE inoculation. The complicated interaction between the immune system and metabolism contributes to the immune responses to SE inoculation of egg-type chickens at 14 dpi at the onset of lay. GC, TNFSF8, CD86, CD274, BLB1 and BLB2 play important roles in response to SE inoculation. The results from this study will deepen the current understanding of the genetic response of the egg-type chicken to SE inoculation at the onset of egg laying.
Collapse
Affiliation(s)
- Guixian Wu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Liying Liu
- College of Life Science, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Yukai Qi
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Yu Sun
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Ning Yang
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guiyun Xu
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Xianyao Li
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, China.,Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, Shandong, 271018, China
| |
Collapse
|
23
|
Nakajima A, Masaki Y, Nakamura T, Kawanami T, Ishigaki Y, Takegami T, Kawano M, Yamada K, Tsukamoto N, Matsui S, Saeki T, Okazaki K, Kamisawa T, Miyashita T, Yakushijin Y, Fujikawa K, Yamamoto M, Hamano H, Origuchi T, Hirata S, Tsuboi H, Sumida T, Morimoto H, Sato T, Iwao H, Miki M, Sakai T, Fujita Y, Tanaka M, Fukushima T, Okazaki T, Umehara H. Decreased Expression of Innate Immunity-Related Genes in Peripheral Blood Mononuclear Cells from Patients with IgG4-Related Disease. PLoS One 2015; 10:e0126582. [PMID: 25973893 PMCID: PMC4431830 DOI: 10.1371/journal.pone.0126582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 04/06/2015] [Indexed: 12/24/2022] Open
Abstract
Background IgG4-related disease (IgG4-RD) is a new clinical entity of unknown etiology characterized by elevated serum IgG4 and tissue infiltration by IgG4-positive plasma cells. Although aberrancies in acquired immune system functions, including increases in Th2 and Treg cytokines observed in patients with IgG4-RD, its true etiology remains unclear. To investigate the pathogenesis of IgG4-RD, this study compared the expression of genes related to innate immunity in patients with IgG4-RD and healthy controls. Materials and Methods Peripheral blood mononuclear cells (PBMCs) were obtained from patients with IgG4-RD before and after steroid therapy and from healthy controls. Total RNA was extracted and DNA microarray analysis was performed in two IgG4-RD patients to screen for genes showing changes in expression. Candidate genes were validated by real-time RT-PCR in 27 patients with IgG4-RD and 13 healthy controls. Results DNA microarray analysis identified 21 genes that showed a greater than 3-fold difference in expression between IgG4-RD patients and healthy controls and 30 genes that showed a greater than 3-fold change in IgG4-RD patients following steroid therapy. Candidate genes related to innate immunity, including those encoding Charcot–Leyden crystal protein (CLC), membrane-spanning 4-domain subfamily A member 3 (MS4A3), defensin alpha (DEFA) 3 and 4, and interleukin-8 receptors (IL8R), were validated by real-time RT-PCR. Expression of all genes was significantly lower in IgG4-RD patients than in healthy controls. Steroid therapy significantly increased the expression of DEFA3, DEFA4 and MS4A3, but had no effect on the expression of CLC, IL8RA and IL8RB. Conclusions The expression of genes related to allergy or innate immunity, including CLC, MS4A3, DEFA3, DEFA4, IL8RA and IL8RB, was lower in PBMCs from patients with IgG4-RD than from healthy controls. Although there is the limitation in the number of patients applied in DNA microarray, impaired expression of genes related to innate immunity may be involved in the pathogenesis of IgG4-RD as well as in abnormalities of acquired immunity.
Collapse
Affiliation(s)
- Akio Nakajima
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Yasufumi Masaki
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Takuji Nakamura
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Takafumi Kawanami
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Tsutomu Takegami
- Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Hospital, Ishikawa 920-8641, Japan
| | - Kazunori Yamada
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Hospital, Ishikawa 920-8641, Japan
| | - Norifumi Tsukamoto
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Shoko Matsui
- Health Administration Center University of Toyama, Toyama 930-0194, Japan
| | - Takako Saeki
- Department of Internal Medicine, Nagaoka Red Cross Hospital, Niigata 940-2085, Japan
| | - Kazuichi Okazaki
- Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Osaka 573-1191, Japan
| | - Terumi Kamisawa
- Department of Internal Medicine, Tokyo Metropolitan Komagome Hospital, Tokyo 113-8677, Japan
| | - Taiichiro Miyashita
- Department of Rheumatology, National Hospital Organization Nagasaki Medical center, Nagasaki 380-8582, Japan
| | - Yoshihiro Yakushijin
- Department of Clinical Oncology, Ehime Graduate School of Medicine, Ehime 791-0295, Japan
| | - Keita Fujikawa
- Department of Rheumatology, Japan Community Healthcare Organization, Isahaya General Hospital, Nagasaki 854-8501, Japan
| | - Motohisa Yamamoto
- Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Hokkaido 060-8543, Japan
| | - Hideaki Hamano
- Medical Informatics Division and Department of Internal Medicine, Gastroenterology, Shinshu University School Hospital, Nagano 390-8621, Japan
| | - Tomoki Origuchi
- First Department of Internal Medicine, Department of Immunology and Rheumatology, Nagasaki Graduate School of Health Sciences, Nagasaki 852-8520, Japan
| | - Shintaro Hirata
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka 807-8555, Japan
| | - Hiroto Tsuboi
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Hisanori Morimoto
- Division of Nephrology, Mitoyo General Hospital, Kagawa 769-1695, Japan
| | - Tomomi Sato
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Haruka Iwao
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Miyuki Miki
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Tomoyuki Sakai
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Yoshimasa Fujita
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Masao Tanaka
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Toshihiro Fukushima
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Toshiro Okazaki
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Hisanori Umehara
- Hematology and Immunology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan; Department of Clinical Immunology, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
24
|
Susceptibility to Salmonella carrier-state: a possible Th2 response in susceptible chicks. Vet Immunol Immunopathol 2014; 159:16-28. [PMID: 24694400 DOI: 10.1016/j.vetimm.2014.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 01/13/2014] [Accepted: 03/02/2014] [Indexed: 11/23/2022]
Abstract
Infection of chicken with Salmonella may lead to a carrier-state characterized by the persistence of bacteria in the ceca for a long period of time and result in their excretion in feces. This excretion is the source of contamination of their congeners and food. During infection, enterocytes are the primary target cells for Salmonella, the producers of soluble factors which launch immune response and cells which are reciprocally responsive to surrounding immune cells. This study used microarrays to compare the gene expression profile during carrier-state of enterocytes purified from infected and control chicks which are either resistant or susceptible to Salmonella Enteritidis carrier-state. In total, we identified 271 genes significantly differentially expressed with an absolute fold change greater than 1.5. A global analysis determined interaction networks between differentially regulated genes. Using an a priori approach, our analyses focused on differentially expressed genes which were transcriptionally linked to cytokines playing a major role in the fate of the immune response. The expression of genes transcriptionally linked to type I interferon and TGF-β was down-regulated in infected chicks from both lines. Gene expression linked to the Th1 axis suggests the latter is inhibited in both lines. Finally, the expression of genes linked to IL-4, IL-5 and IL-13 indicates that susceptibility to carrier-state could be associated with a Th2 bias. Overall, these results highlight that the response to Salmonella during the acute phase and carrier-state is different and that enterocytes play a central role in this response.
Collapse
|
25
|
Eisenreich W, Heesemann J, Rudel T, Goebel W. Metabolic host responses to infection by intracellular bacterial pathogens. Front Cell Infect Microbiol 2013; 3:24. [PMID: 23847769 PMCID: PMC3705551 DOI: 10.3389/fcimb.2013.00024] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
The interaction of bacterial pathogens with mammalian hosts leads to a variety of physiological responses of the interacting partners aimed at an adaptation to the new situation. These responses include multiple metabolic changes in the affected host cells which are most obvious when the pathogen replicates within host cells as in case of intracellular bacterial pathogens. While the pathogen tries to deprive nutrients from the host cell, the host cell in return takes various metabolic countermeasures against the nutrient theft. During this conflicting interaction, the pathogen triggers metabolic host cell responses by means of common cell envelope components and specific virulence-associated factors. These host reactions generally promote replication of the pathogen. There is growing evidence that pathogen-specific factors may interfere in different ways with the complex regulatory network that controls the carbon and nitrogen metabolism of mammalian cells. The host cell defense answers include general metabolic reactions, like the generation of oxygen- and/or nitrogen-reactive species, and more specific measures aimed to prevent access to essential nutrients for the respective pathogen. Accurate results on metabolic host cell responses are often hampered by the use of cancer cell lines that already exhibit various de-regulated reactions in the primary carbon metabolism. Hence, there is an urgent need for cellular models that more closely reflect the in vivo infection conditions. The exact knowledge of the metabolic host cell responses may provide new interesting concepts for antibacterial therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Lehrstuhl für Biochemie, Center of Isotopologue Profiling, Technische Universität München Garching, Germany
| | | | | | | |
Collapse
|
26
|
Hulst M, Smits M, Vastenhouw S, de Wit A, Niewold T, van der Meulen J. Transcription networks responsible for early regulation of Salmonella-induced inflammation in the jejunum of pigs. JOURNAL OF INFLAMMATION-LONDON 2013; 10:18. [PMID: 23590759 PMCID: PMC3637394 DOI: 10.1186/1476-9255-10-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/10/2013] [Indexed: 05/16/2023]
Abstract
Background The aim of this study was to identify transcription factors/regulators that play a crucial role in steering the (innate) immune response shortly (within a few hours) after the first contact of the intestinal mucosa with an inflammatory mediator, and to test whether the processes regulated by these factors/regulators can be modulated by chemical substances of natural origin. Methods We experimentally induced inflammation by perfusion of surgically applied jejunal loops with Salmonella enterica subspecies enterica serovar Typhimurium DT104 in three pigs. Segments of mock and Salmonella treated loops were dissected after 2, 4 and 8 hours of perfusion. IL8 and IL1-beta mRNA expression levels were measured in mucosal scrapings of all segments. Furthermore, intra-animal microarray comparisons (isogenic) between Salmonella and mock treated segments after 8 hours, and inter-animal comparisons between similar Salmonella-treated loops of each pig at 2 and 4 hours, were performed. Results IL-1beta and IL8 mRNA levels, and intra-animal microarray comparisons at 8 hours between Salmonella and mock treated segments showed that the response-time and type of response to Salmonella was different in all three pigs. This plasticity allowed us to extract a comprehensive set of differentially expressed genes from inter-animal comparisons at 2 and 4 hours. Pathway analysis indicated that many of these genes play a role in induction and/or tempering the inflammatory response in the intestine. Among them a set of transcription factors/regulators known to be involved in regulation of inflammation, but also factors/regulators for which involvement was not expected. Nine out of twenty compounds of natural origin, which according to literature had the potential to modulate the activity of these factors/regulators, were able to stimulate or inhibit a Salmonella-induced mRNA response of inflammatory-reporter genes IL8 and/or nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha in cultured intestinal porcine epithelial cells. Conclusions We describe a set of transcription factors/regulators possibly involved in regulation of “very early” immune mechanism which determines the inflammatory status of the intestine later on. In addition, we show that these mechanisms may be modulated by chemical substances of natural origin.
Collapse
Affiliation(s)
- Marcel Hulst
- Livestock Research of Wageningen University and Research Centre, P,O, Box 65, Lelystad, 8200AB, The Netherlands.
| | | | | | | | | | | |
Collapse
|
27
|
Martins RP, Collado-Romero M, Arce C, Lucena C, Carvajal A, Garrido JJ. Exploring the immune response of porcine mesenteric lymph nodes to Salmonella enterica serovar Typhimurium: an analysis of transcriptional changes, morphological alterations and pathogen burden. Comp Immunol Microbiol Infect Dis 2012; 36:149-60. [PMID: 23274115 DOI: 10.1016/j.cimid.2012.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 11/14/2012] [Accepted: 11/19/2012] [Indexed: 12/22/2022]
Abstract
Infections caused by Salmonella enterica serovar Typhimurium (S. typhimurium) cause important economic problems in the swine industry and threaten the integrity of a safe and healthy food supply. Controlling the prevalence of Salmonella in pig production requires a thorough knowledge of the response processes that occurs in the gut associated immune tissues. To explore the in vivo porcine response to S. typhimurium, MLN samples from four control pigs and twelve infected animals at 1, 2 and 6 days post infection (dpi) were collected to quantify the mRNA expression of gene coding for 42 innate immune-related molecules. In addition, the presence of S. typhimurium in MLN was examined and its effect on tissue micro-anatomy. Higher S. typhimurium loads were observed at 2dpi, triggering an innate immune response, marked by a substantial infiltration of phagocytes and up-regulation of pro-inflammatory genes. Such response resulted in a significant decrease in pathogen burden in MLN at 6dpi, although Salmonella could not be completely eliminated from tissue. Furthermore, our results suggest that in porcine infections, S. typhimurium might interferes with dendritic cell-T cell interactions and this strategy could be involved in the conversion of Salmonella infected pigs to a carrier state.
Collapse
Affiliation(s)
- Rodrigo Prado Martins
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Universidad de Córdoba, Campus de Rabanales, Edificio Gregor Mendel C5, 14071 Córdoba, Spain
| | | | | | | | | | | |
Collapse
|
28
|
Li C, Zhang Y, Wang R, Lu J, Nandi S, Mohanty S, Terhune J, Liu Z, Peatman E. RNA-seq analysis of mucosal immune responses reveals signatures of intestinal barrier disruption and pathogen entry following Edwardsiella ictaluri infection in channel catfish, Ictalurus punctatus. FISH & SHELLFISH IMMUNOLOGY 2012; 32:816-827. [PMID: 22366064 DOI: 10.1016/j.fsi.2012.02.004] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/02/2012] [Accepted: 02/03/2012] [Indexed: 05/31/2023]
Abstract
The mucosal surfaces of fish (gill, skin, gastrointestinal tract) are important sites of bacterial exposure and host defense mechanisms. In mammalian systems, the intestinal epithelium is well characterized as both a selectively permeable barrier regulated by junctional proteins and as a primary site of infection for a number of enteric pathogens including viruses, bacteria, and parasites. The causative bacterium of enteric septicemia of catfish, Edwardsiella ictaluri, is believed to gain entry through the intestinal epithelium, with previous research using a rat intestinal epithelial cell line (IEC-6) indicating actin polymerization and receptor-mediated endocytosis as potential mechanisms of uptake. Here, we utilized high-throughput RNA-seq to characterize the role of the intestinal epithelial barrier following E. ictaluri challenge. A total of 197.6 million reads were obtained and assembled into 176,481 contigs with an average length of 893.7 bp and N50 of 1676 bp. The assembled contigs contained 14,457 known unigenes, including 2719 genes not previously identified in other catfish transcriptome studies. Comparison of digital gene expression between challenged and control samples revealed 1633 differentially expressed genes at 3 h, 24 h, and 3 day following exposure. Gene pathway analysis of the differentially expressed gene set indicated the centrality of actin cytoskeletal polymerization/remodelling and junctional regulation in pathogen entry and subsequent inflammatory responses. The expression patterns of fifteen differentially expressed genes related to intestinal epithelial barrier dysfunction were validated by quantitative real-time RT-PCR (average correlation coeff. 0.92, p < 0.001). Our results set a foundation for future studies comparing mechanisms of pathogen entry and mucosal immunity across several important catfish pathogens including E. ictaluri, Edwardsiellatarda, Flavobacterium columnare, and virulent atypical Aeromonas hydrophila. Understanding of molecular mechanisms of pathogen entry during infection will provide insight into strategies for selection of resistant catfish brood stocks against various diseases.
Collapse
Affiliation(s)
- Chao Li
- Department of Fisheries and Allied Aquacultures, Aquatic Genomics Unit, Auburn University, Auburn, AL 36849, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Integrating signals from the ECM (extracellular matrix) via the cell surface into the nucleus is an essential feature of multicellular life and often malfunctions in cancer. To date many signal transducers known as shuttle proteins have been identified that act as both: a cytoskeletal and a signalling protein. Here, we highlight the interesting member of the Zyxin family TRIP6 [thyroid receptor interactor protein 6; also designated ZRP-1 (zyxin-related protein 1)] and review current literature to define its role in cell physiology and cancer. TRIP6 is a versatile scaffolding protein at FAs (focal adhesions) involved in cytoskeletal organization, coordinated cell migration and tissue invasion. Via its LIM and TDC domains TRIP6 interacts with different components of the LPA (lysophosphatidic acid), NF-κB (nuclear factor κB), glucocorticoid and AMPK (AMP-activated protein kinase) signalling pathway and thereby modulates their activity. Within the nucleus TRIP6 acts as a transcriptional cofactor regulating the transcriptional responses of these pathways. Moreover, intranuclear TRIP6 associates with proteins ensuring telomere protection and hence may contribute to genome stability. Accordingly, TRIP6 is engaged in key cellular processes such as cell proliferation, differentiation and survival. These diverse functions of TRIP6 are found to be dysregulated in various cancers and may have pleiotropic roles in tumour initiation, tumour growth and metastasis, which turn TRIP6 into an attractive candidate for cancer diagnosis and targeted therapy.
Collapse
|
30
|
Grasberger H, De Deken X, Mayo OB, Raad H, Weiss M, Liao XH, Refetoff S. Mice deficient in dual oxidase maturation factors are severely hypothyroid. Mol Endocrinol 2012; 26:481-92. [PMID: 22301785 DOI: 10.1210/me.2011-1320] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dual oxidases (DUOX1 and DUOX2) are evolutionary conserved reduced nicotinamide adenine dinucleotide phosphate oxidases responsible for regulated hydrogen peroxide (H(2)O(2)) release of epithelial cells. Specific maturation factors (DUOXA1 and DUOXA2) are required for targeting of functional DUOX enzymes to the cell surface. Mutations in the single-copy Duox and Duoxa genes of invertebrates cause developmental defects with reduced survival, whereas knockdown in later life impairs intestinal epithelial immune homeostasis. In humans, mutations in both DUOX2 and DUOXA2 can cause congenital hypothyroidism with partial iodide organification defects compatible with a role of DUOX2-generated H(2)O(2) in driving thyroid peroxidase activity. The DUOX1/DUOXA1 system may account for residual iodide organification in patients with loss of DUOX2, but its physiological function is less clear. To provide a murine model recapitulating complete DUOX deficiency, we simultaneously targeted both Duoxa genes by homologous recombination. Knockout of Duoxa genes (Duoxa(-/-) mice) led to a maturation defect of DUOX proteins lacking Golgi processing of N-glycans and to loss of H(2)O(2) release from thyroid tissue. Postnatally, Duoxa(-/-) mice developed severe goitreous congenital hypothyroidism with undetectable serum T4 and maximally disinhibited TSH levels. Heterozygous mice had normal thyroid function parameters. (125)I uptake and discharge studies and probing of iodinated TG epitopes corroborated the iodide organification defect in Duoxa(-/-) mice. Duoxa(-/-) mice on continuous T4 replacement from P6 showed normal growth without an overt phenotype. Our results confirm in vivo the requirement of DUOXA for functional expression of DUOX-based reduced nicotinamide adenine dinucleotide phosphate oxidases and the role of DUOX isoenzymes as sole source of hormonogenic H(2)O(2).
Collapse
Affiliation(s)
- Helmut Grasberger
- Biomedical Science Research Building, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Richter-Dahlfors A, Rhen M, Udekwu K. Tissue microbiology provides a coherent picture of infection. Curr Opin Microbiol 2012; 15:15-22. [DOI: 10.1016/j.mib.2011.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 12/26/2022]
|
32
|
Medina C, Santero E, Gómez-Skarmeta JL, Royo JL. Engineered Salmonella allows real-time heterologous gene expression monitoring within infected zebrafish embryos. J Biotechnol 2011; 157:413-6. [PMID: 22178780 DOI: 10.1016/j.jbiotec.2011.11.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 11/28/2011] [Indexed: 01/26/2023]
Abstract
Microbial host-pathogen interactions have been traditionally well studied at genetic and physiological levels, but cell-resolution analyses have been particularly scarce. This has been especially remarkable for intracellular parasites for two major reasons: first, the inherent loss of bacteria traceability once infects its hosts; second and more important, the limited availability of genetic tools that allow a tight regulated expression of bacterial virulence genes once inside the host tissues. Here we present novel data supporting the use of zebrafish embryos to monitor Salmonella enterica serovar Thyphimurium infection. Intravenous infection of Salmonella can be easily monitored using in vivo fluorescence that allows the visualization of free-swimming bacteria through the circulatory system. Moreover, we have engineered Salmonella to voluntarily activate heterologous gene expression at any point during infection once inside the zebrafish macrophages using a salicylate-based expression system. This approach allows real-time cell-resolution in vivo monitoring of the infection. All together, this approach paves the road to cell-based resolution experiments that would be harder to mimic in other vertebrate infection models.
Collapse
Affiliation(s)
- Carlos Medina
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/Consejo Superior de Investigaciones Científicas/Junta de Andalucía, Ctra. Utrera, Km 1. 41013-Sevilla, Spain
| | | | | | | |
Collapse
|
33
|
Small intestinal intraepithelial lymphocytes expressing CD8 and T cell receptor γδ are involved in bacterial clearance during Salmonella enterica serovar Typhimurium infection. Infect Immun 2011; 80:565-74. [PMID: 22144492 DOI: 10.1128/iai.05078-11] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The intestinal immune system is crucial for the maintenance of mucosal homeostasis and has evolved under the dual pressure of protecting the host from pathogenic infection and coexisting with the dense and diverse commensal organisms in the lumen. Intestinal intraepithelial lymphocytes (iIELs) are the first element of the host T cell compartment available to respond to oral infection by pathogens. This study demonstrated that oral infection by Salmonella enterica serovar Typhimurium promoted the expansion of iIELs, particularly CD8(+) TCRγδ(+) IELs, enhanced expression of NKG2D on iIELs, increased expression of MULT1, and decreased expression of Qa-1 by intestinal epithelial cells (IECs), leading to activation of, particularly, CD8(+) TCRγδ(+) iIELs and cytolytic activity against S. Typhimurium-infected IECs. Blockade of NKG2D recognition or depletion of TCRγδ(+) cells using a depleting monoclonal antibody significantly attenuated the clearance of S. Typhimurium in the intestine and other tissues. This study suggests that iIELs, particularly CD8(+) TCRγδ(+) iIELs, play important roles in the detection of pathogenic bacteria and eradication of infected epithelial cells and, thus, provide protection against invading pathogens. These data further our understanding of the mechanisms by which the immune system of the intestinal mucosa discriminates between pathogenic and commensal organisms.
Collapse
|
34
|
Liu X, Wu S, Xia Y, Li XE, Xia Y, Zhou ZD, Sun J. Wingless homolog Wnt11 suppresses bacterial invasion and inflammation in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2011; 301:G992-G1003. [PMID: 21903761 PMCID: PMC3233790 DOI: 10.1152/ajpgi.00080.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Wnt11 plays an essential role in gastrointestinal epithelial proliferation, and previous investigations have focused on development and immune responses. However, the roles of how enteric bacteria regulate Wnt11 and how Wnt11 modulates the host response to pathogenic bacteria remain unexplored. This study investigated the effects of Salmonella infection on Wnt activation in intestinal epithelial cells. We found that Wnt11 mRNA and protein expression were elevated after Salmonella colonization. Wnt11 protein secretion in epithelial cells was also elevated after bacterial infection. Furthermore, we demonstrated that pathogenic Salmonella regulated Wnt11 expression and localization in vivo. We found a decrease in Salmonella invasion in cells with Wnt11 overexpression compared with cells with normal Wnt11 level. IL-8 mRNA in Wnt11-transfected cells was low; however, it was enhanced in cells with a low level of Wnt11 expression. Functionally, Wnt11 overexpression inhibited Salmonella-induced apoptosis. AvrA is a known bacterial effector protein that stabilizes β-catenin, the downstream regulator of Wnt signaling, and inhibits bacterially induced intestinal inflammation. We observed that Wnt11 expression, secretion, and transcriptional activity were regulated by Salmonella AvrA. Overall, Wnt11 is involved in the protection of the host intestinal cells by blocking the invasion of pathogenic bacteria, suppressing inflammation, and inhibiting apoptosis. Wnt11 is a novel and important contributor to intestinal homeostasis and host defense.
Collapse
Affiliation(s)
- Xingyin Liu
- 1Gastroenterology & Hepatology Division, Department of Medicine,
| | - Shaoping Wu
- 1Gastroenterology & Hepatology Division, Department of Medicine,
| | - Yinglin Xia
- 4Department of Biostatistics and Computational Biology, and
| | - Xi Emma Li
- 1Gastroenterology & Hepatology Division, Department of Medicine,
| | - Yuxuan Xia
- 5Department of Pathology, University of Rochester, Rochester, New York; and ,6Brighton High School, Rochester, New York
| | | | - Jun Sun
- 1Gastroenterology & Hepatology Division, Department of Medicine, ,2Department of Microbiology and Immunology, ,3Wilmot Cancer Center,
| |
Collapse
|
35
|
Probiotics, nuclear receptor signaling, and anti-inflammatory pathways. Gastroenterol Res Pract 2011; 2011:971938. [PMID: 21808643 PMCID: PMC3144714 DOI: 10.1155/2011/971938] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/28/2011] [Accepted: 05/19/2011] [Indexed: 02/07/2023] Open
Abstract
There is increased investigation of the human microbiome as it relates to health and disease. Dysbiosis is implicated in various clinical conditions including inflammatory bowel disease (IBD). Probiotics have been explored as a potential treatment for IBD and other diseases. The mechanism of action for probiotics has yet to be fully elucidated. This paper discusses novel mechanisms of action for probiotics involving anti-inflammatory signaling pathways. We highlight recent progress in probiotics and nuclear receptor signaling, such as peroxisome-proliferator-activated receptor gamma (PPARγ) and vitamin D receptor (VDR). We also discuss future areas of investigation.
Collapse
|
36
|
Zhou C, Pridgen B, King N, Xu J, Breslow JL. Hyperglycemic Ins2AkitaLdlr⁻/⁻ mice show severely elevated lipid levels and increased atherosclerosis: a model of type 1 diabetic macrovascular disease. J Lipid Res 2011; 52:1483-93. [PMID: 21606463 DOI: 10.1194/jlr.m014092] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Accelerated atherosclerosis is the leading cause of death in type 1 diabetes, but the mechanism of type 1 diabetes-accelerated atherosclerosis is not well understood, in part due to the lack of a good animal model for the long-term studies required. In an attempt to create a model for studying diabetic macrovascular disease, we have generated type 1 diabetic Akita mice lacking the low density lipoprotein receptor (Ins2(Akita)Ldlr⁻/⁻). Ins2(Akita)Ldlr⁻/⁻ mice were severely hyperglycemic with impaired glucose tolerance. Compared with Ldlr⁻/⁻ mice, 20-week-old Ins2(Akita)Ldlr⁻/⁻ mice fed a 0.02% cholesterol AIN76a diet showed increased plasma triglyceride and cholesterol levels, and increased aortic root cross-sectional atherosclerotic lesion area [224% (P < 0.001) in males and 30% (P < 0.05) in females]. Microarray and quantitative PCR analyses of livers from Ins2(Akita)Ldlr⁻/⁻ mice revealed altered expression of lipid homeostatic genes, including sterol-regulatory element binding protein (Srebp)1, liver X receptor (Lxr)α, Abca1, Cyp7b1, Cyp27a1, and Lpl, along with increased expression of pro-inflammatory cytokine genes, including interleukin (Il)1α, Il1β, Il2, tumor necrosis factor (Tnf)α, and Mcp1. Immunofluorescence staining showed that the expression levels of Mcp1, Tnfα, and Il1β were also increased in the atherosclerotic lesions and artery walls of Ins2(Akita)Ldlr⁻/⁻ mice. Thus, the Ins2(Akita)Ldlr⁻/⁻ mouse appears to be a promising model for mechanistic studies of type 1 diabetes-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Changcheng Zhou
- Laboratory of Biochemical Genetics and Metabolism, Rockefeller University, New York, NY, USA
| | | | | | | | | |
Collapse
|
37
|
Wu S, Sun J. Vitamin D, vitamin D receptor, and macroautophagy in inflammation and infection. DISCOVERY MEDICINE 2011; 11:325-335. [PMID: 21524386 PMCID: PMC3285235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Vitamin D is involved in mineral and bone homeostasis, immune responses, anti-inflammation, anti-infection, and cancer prevention. Vitamin D receptor (VDR) is a nuclear receptor that mediates most biological functions of 1,25(OH)(2)D(3) or vitamin D(3), the active form of vitamin D. Recently, vitamin D(3)-induced autophagy has been reported. Autophagy is a lysosome-mediated catabolic pathway classified into three different types: macroautophagy, microautophagy, and chaperone-mediated autophagy. Autophagy contributes to anti-aging, antimicrobial defense, and tumor suppression. The functions of autophagy overlap remarkably with those of vitamin D/VDR signaling. This review focuses on vitamin D(3), VDR, and macroautophagy in inflammation and infection. We place emphasis on the regulatory roles of vitamin D(3) on autophagy at different steps, including induction, nucleation, elongation to maturation, and degradation. We summarize the known molecular mechanisms of vitamin D/VDR signaling on autophagy homeostasis. The potential application of the insights gleaned from these research findings to anti-inflammation and anti-infection is also discussed.
Collapse
Affiliation(s)
- Shaoping Wu
- Department of Medicine, University of Rochester, New York 14642, USA
| | | |
Collapse
|
38
|
Liu X, Lu R, Xia Y, Wu S, Sun J. Eukaryotic signaling pathways targeted by Salmonella effector protein AvrA in intestinal infection in vivo. BMC Microbiol 2010; 10:326. [PMID: 21182782 PMCID: PMC3027599 DOI: 10.1186/1471-2180-10-326] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 12/23/2010] [Indexed: 11/22/2022] Open
Abstract
Background The Salmonella AvrA gene is present in 80% of Salmonella enterica serovar strains. AvrA protein mimics the activities of some eukaryotic proteins and uses these activities to the pathogen's advantage by debilitating the target cells, such as intestinal epithelial cells. Therefore, it is important to understand how AvrA works in targeting eukaryotic signaling pathways in intestinal infection in vivo. In this study, we hypothesized that AvrA interacts with multiple stress pathways in eukaryotic cells to manipulate the host defense system. A whole genome approach combined with bioinformatics assays was used to investigate the in vivo genetic responses of the mouse colon to Salmonella with or without AvrA protein expression in the early stage (8 hours) and late stage (4 days). Specifically, we examined the gene expression profiles in mouse colon as it responded to pathogenic Salmonella stain SL1344 (with AvrA expression) or SB1117 (without AvrA expression). Results We identified the eukaryotic targets of AvrA and the cell signaling pathways regulated by AvrA in vivo. We found that pathways, such as mTOR, NF-kappaB, platelet-derived growth factors, vascular endothelial growth factor, oxidative phosphorylation, and mitogen-activated protein kinase signaling are specifically regulated by AvrA in vivo and are associated with inflammation, anti-apoptosis, and proliferation. At the early stage of Salmonella infection, AvrA mainly targeted pathways related to nuclear receptor signaling and oxidative phosphorylation. At the late stage of Salmonella infection, AvrA is associated with interferon-gamma responses. Conclusion Both early and late phases of the host response exhibit remarkable specificity for the AvrA+ Salmonella. Our studies provide new insights into the eukaryotic molecular cascade that combats Salmonella-associated intestinal infection in vivo.
Collapse
Affiliation(s)
- Xingyin Liu
- Department of Medicine, Gastroenterology & Hepatology Division, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|