1
|
Guo G, Zhang Y, Wei D, Wang Z, Li Q, Yu Y, Zhang W. Contribution of nadR to the cell growth and virulence of Streptococcus suis serotype 2. Vet Microbiol 2024; 288:109928. [PMID: 38056180 DOI: 10.1016/j.vetmic.2023.109928] [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: 02/08/2022] [Revised: 10/06/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Streptococcus suis serotype 2 (SS2) has been reported to be a highly invasive pathogen in swine and a zoonotic agent for humans. Although many bacterial virulence factors have been identified, our an insightful understanding of SS2 pathogenicity is lacking. The gene nadR, encoding nicotinamide-nucleotide adenylyltransferase, was first reported as a regulator and transporter of the intracellular NAD synthesis pathway in Salmonella typhimurium. In this study, we constructed a mutant strain of nadR (ΔnadR) to test the phenotypic and virulence variations between the deletion mutant and the wild-type strain ZY05719. The phenotypic experimental results showed that ΔnadR obviously inhibited cell growth and exhibited shorter chains than WT. The growth defect of ΔnadR was caused by the loss of the function of nadR for transporting the substrates nicotinamide mononucleotide and nicotinamide riboside in the intracellular NAD synthesis pathway. In the process of interaction with the host, ΔnadR participated in adherence and invasion to the host cells, and it was more easily cleared by RAW264.7 macrophages. More importantly, both zebrafish and BALB/c mice in vivo virulence experimental results showed that ΔnadR dramatically attenuated the virulence of SS2, and the ability of ΔnadR to colonize tissues was notably reduced in comparison with that of WT in the BALB/c mice infection model. To the best of our knowledge, this is the first time to demonstrate that nadR not only plays an important role in bacterial growth, but also in connection with the virulence of SS2 as a global transcriptional regulator.
Collapse
Affiliation(s)
- Genglin Guo
- Shandong Institute of Sericulture, Shandong Academy of Agricultural Sciences, Yantai, China
| | - Yuhang Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China; The Sanya Institute of Nanjing Agricultural University, Sanya, China
| | - Dan Wei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China; The Sanya Institute of Nanjing Agricultural University, Sanya, China
| | - Zhuohao Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China; The Sanya Institute of Nanjing Agricultural University, Sanya, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yanfei Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture and Rural Affairs, Nanjing, China; School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China; The Sanya Institute of Nanjing Agricultural University, Sanya, China.
| |
Collapse
|
2
|
Wang J, Liang P, Sun H, Wu Z, Gottschalk M, Qi K, Zheng H. Comparative transcriptomic analysis reveal genes involved in the pathogenicity increase of Streptococcus suis epidemic strains. Virulence 2022; 13:1455-1470. [PMID: 36031944 PMCID: PMC9423846 DOI: 10.1080/21505594.2022.2116160] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Streptococcus suis epidemic strains were responsible for two outbreaks in China and possessed increased pathogenicity which was featured prominently by inducing an excessive inflammatory response at the early phase of infection. To discover the critical genes responsible for the pathogenicity increase of S. suis epidemic strains, the genome-wide transcriptional profiles of epidemic strain SC84 were investigated at the early phase of interaction with BV2 cells. The overall low expression levels of 89K pathogenicity island (PAI) and 129 known virulence genes in the SC84 interaction groups indicated that its pathogenicity increase should be attributed to novel mechanisms. Using highly pathogenic strain P1/7 and intermediately pathogenic strain 89–1591 as controls, 11 pathogenicity increase crucial genes (PICGs) and 38 pathogenicity increase-related genes (PIRGs) were identified in the SC84 incubation groups. The PICGs encoded proteins related to the methionine biosynthesis/uptake pathway and played critical roles in the pathogenicity increase of epidemic strains. A high proportion of PIRGs encoded surface proteins related to host cell adherence and immune escape, which may be conducive to the pathogenicity increase of epidemic strains by rapidly initiating infection. The fact that none of PICGs and PIRGs belonged to epidemic strain-specific gene indicated that the pathogenicity increase of epidemic strain may be determined by the expression level of genes, rather than the presence of them. Our results deepened the understanding on the mechanism of the pathogenicity increase of S. suis epidemic strains and provided novel approaches to control the life-threatening infections of S. suis epidemic strains.
Collapse
Affiliation(s)
- Jianping Wang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Pujun Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
- OIE Reference Lab for Swine Streptococcosis, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hui Sun
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Zongfu Wu
- Swine and Poultry Infectious Diseases Research Center, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Marcelo Gottschalk
- Department of Clinical Laboratory, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Kexin Qi
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| | - Han Zheng
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, China
| |
Collapse
|
3
|
Identification of Proteins Responsible for High Activity of Cysteine Proteinase Inhibitor in the Blood of Nile Tilapia Oreochromis niloticus. FISHES 2022. [DOI: 10.3390/fishes7040161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cysteine proteinase inhibitors (CPIs) protect tissues and organs against cysteine proteinases in animal blood and have attracted much attention for use in food processing and medical sciences for humans and animals. Several CPI proteins, which include stefins, cystatins, kininogens, histidine-rich glycoproteins (HRG) and fetuins, have been identified and characterized in mammals. Fish blood also contains high CPI activity, but the identity of the major protein responsible for this activity has not been clarified. This study was conducted to screen CPI activity by examining papain inhibitory activity from various different tissues in Nile tilapia Oreochromis niloticus and to identify major proteins for the activity in the blood. CPI activity was highest in the serum among the tissues screened in this study (at least fourfold higher than in other tissues)(P < 0.05). Major proteins for CPI activity in serum were purified using a CNBr-activated sepharose 4B column, gel filtration and an ion exchange FPLC column. From these purifications, two proteins with strong CPI activity were isolated and partially sequenced. Based on their molecular weights and partial amino sequences, the two major proteins with CPI activity from the blood in this species were found to be fetuin B (60 kDa) and kininogen (54 kDa).
Collapse
|
4
|
Fan J, Zhao L, Hu Q, Li S, Li H, Zhang Q, Zou G, Zhang L, Li L, Huang Q, Zhou R. Screening for Virulence-Related Genes via a Transposon Mutant Library of Streptococcus suis Serotype 2 Using a Galleria mellonella Larvae Infection Model. Microorganisms 2022; 10:microorganisms10050868. [PMID: 35630313 PMCID: PMC9143085 DOI: 10.3390/microorganisms10050868] [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: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 01/27/2023] Open
Abstract
Streptococcus suis (S. suis) is a zoonotic bacterial pathogen causing lethal infections in pigs and humans. Identification of virulence-related genes (VRGs) is of great importance in understanding the pathobiology of a bacterial pathogen. To identify novel VRGs, a transposon (Tn) mutant library of S. suis strain SC19 was constructed in this study. The insertion sites of approximately 1700 mutants were identified by Tn-seq, which involved 417 different genes. A total of 32 attenuated strains were identified from the library by using a Galleria mellonella larvae infection model, and 30 novel VRGs were discovered, including transcription regulators, transporters, hypothetical proteins, etc. An isogenic deletion mutant of hxtR gene (ΔhxtR) and its complementary strain (CΔhxtR) were constructed, and their virulence was compared with the wild-type strain in G. mellonella larvae and mice, which showed that disruption of hxtR significantly attenuated the virulence. Moreover, the ΔhxtR strain displayed a reduced survival ability in whole blood, increased sensitivity to phagocytosis, increased chain length, and growth defect. Taken together, this study performed a high throughput screening for VRGs of S. suis using a G. mellonella larvae model and further characterized a novel critical virulence factor.
Collapse
Affiliation(s)
- Jingyan Fan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Lelin Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Siqi Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Qianqian Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Geng Zou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Liangsheng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
- Correspondence: (Q.H.); (R.Z.)
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.F.); (L.Z.); (Q.H.); (S.L.); (H.L.); (Q.Z.); (G.Z.); (L.Z.); (L.L.)
- International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan 430070, China
- The HZAU-HVSEN Research Institute, Wuhan 430042, China
- Correspondence: (Q.H.); (R.Z.)
| |
Collapse
|
5
|
Essfeld F, Reinwald H, Salinas G, Schäfers C, Eilebrecht E, Eilebrecht S. Transcriptomic profiling of clobetasol propionate-induced immunosuppression in challenged zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 233:113346. [PMID: 35228030 DOI: 10.1016/j.ecoenv.2022.113346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
In the ecotoxicological hazard assessment of chemicals, the detection of immunotoxicity is currently neglected. This is mainly due to the complexity of the immune system and the consequent lack of standardized procedures and markers for the comprehensive assessment of immunotoxic modes of action. In this study, we present a new approach applying transcriptome profiling to an immune challenge with a mixture of pathogen-associated molecular patterns (PAMPs) in zebrafish embryos, analyzing differential gene expression during acute infection with and without prior exposure to the immunosuppressive drug clobetasol propionate (CP). While PAMP injection itself triggered biological processes associated with immune activation, some of these genes were more differentially expressed upon prior exposure to CP than by immune induction alone, whereas others showed weaker or no differential regulation in response to the PAMP stimulus. All of these genes responding differently to PAMP after prior CP exposure showed additivity of PAMP- and CP-induced effects, indicating independent regulatory mechanisms. The transcriptomic profiles suggest that CP impaired innate immune induction by attenuating the response of genes involved in antigen processing, TLR signaling, NF-КB signaling, and complement activation. We propose this approach as a powerful method for detecting gene biomarkers for immunosuppressive modes of action, as it was able to identify alternatively regulated processes and pathways in a sublethal, acute infection zebrafish embryo model. This allowed to define biomarker candidates for immune-mediated effects and to comprehensively characterize immunosuppression. Ultimately, this work contributes to the development of molecular biomarker-based environmental hazard assessment of chemicals in the future.
Collapse
Affiliation(s)
- Fabian Essfeld
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany; Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Hannes Reinwald
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany; Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Gabriela Salinas
- NGS-Services for Integrative Genomics, University of Göttingen, Göttingen, Germany
| | - Christoph Schäfers
- Department Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Elke Eilebrecht
- Department Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Sebastian Eilebrecht
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany.
| |
Collapse
|
6
|
Genome analysis provides insight into hyper-virulence of Streptococcus suis LSM178, a human strain with a novel sequence type 1005. Sci Rep 2021; 11:23919. [PMID: 34907269 PMCID: PMC8671398 DOI: 10.1038/s41598-021-03370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus suis has been well-recognized as a zoonotic pathogen worldwide, and the diversity and unpredictable adaptive potential of sporadic human strains represent a great risk to the public health. In this study, S. suis LSM178, isolated from a patient in contact with pigs and raw pork, was assessed as a hyper-virulent strain and interpreted for the virulence based on its genetic information. The strain was more invasive for Caco-2 cells than two other S. suis strains, SC19 and P1/7. Sequence analysis designated LSM178 with serotype 2 and a novel sequence type 1005. Phylogenetic analysis showed that LSM178 clustered with highly virulent strains including all human strains and epidemic strains. Compared with other strains, these S. suis have the most and the same virulent factors and a type I-89 K pathogenicity island. Further, groups of genes were identified to distinguish these highly virulent strains from other generally virulent strains, emphasizing the key roles of genes modeling transcription, cell barrier, replication, recombination and repair on virulence regulation. Additionally, LSM178 contains a novel prophage conducive potentially to pathogenicity.
Collapse
|
7
|
Wang X, Sun J, Bian C, Wang J, Liang Z, Shen Y, Yao H, Huang J, Wang L, Zheng H, Wu Z. The population structure, antimicrobial resistance, and pathogenicity of Streptococcus suis cps31. Vet Microbiol 2021; 259:109149. [PMID: 34147764 DOI: 10.1016/j.vetmic.2021.109149] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/05/2021] [Indexed: 12/15/2022]
Abstract
Streptococcus suis is a zoonotic pathogen that can cause invasive infections in humans and pigs. The S. suis cps31 strains (SS31) were frequently isolated from healthy or diseased pigs and one human infection case caused by SS31 was reported in Thailand in 2015. However, except for a few epidemiologic studies, little information is available for SS31. To characterize SS31, a total of 75 SS31 strains were analyzed, including 52 strains that were isolated from healthy or diseased pigs and 23 strains whose information was accessed from NCBI. The MLST analysis showed that SS31 exhibited high heterogeneity. The phylogenetic analysis and minimum core-genome (MCG) classification revealed that 75 strains were clustered into 3 lineages. Strains from NCBI mainly at Lineage 2 belong to MCG7-3, and most of strains from China at Lineage 3 belong to MCG7-2. This finding indicated that their evolutionary path was different. All SS31 strains were resistant to more than three classes of antimicrobial agents, and major antimicrobial resistance genes for strains from Lineage 3 were carried by prophages. This observation is different from the previous observation that integrative conjugative elements and integrative and mobilizable elements are major vehicles of antimicrobial resistance genes for S. suis. In addition to strains isolated from diseased pigs, seven of 47 strains isolated from clinically healthy pigs were also pathogenic in a zebrafish infection model. These findings reveal unique characteristics of SS31 and contribute to establishing public health surveillance for SS31 and clarifying the diversity of S. suis.
Collapse
Affiliation(s)
- Xiaoming Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Junjie Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chen Bian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Jianping Wang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, 102206, China
| | - Zijing Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Yanling Shen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Han Zheng
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, 102206, China.
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| |
Collapse
|
8
|
Guo G, Kong X, Du D, Wei D, Yu Y, Zhang W. Genome-wide association study identifies the virulence-associated marker in Streptococcus suis serotype 2. INFECTION GENETICS AND EVOLUTION 2021; 92:104894. [PMID: 33964473 DOI: 10.1016/j.meegid.2021.104894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022]
Abstract
Streptococcus suis (S. suis) has been reported to be a highly invasive pathogen in swine, which causes severe infections like meningitis, arthritis and septicemia, and also a zoonotic agent for humans. Although many putative virulence factors (VFs) have been identified, the exact and wildly accepted virulence associated marker and pathogenesis mechanism of S. suis are still unclear. To establish connection of the genotypes with virulence phenotypes, we performed an "internal standard" method based on the zebrafish model to assess the virulence phenotypes of S. suis and did the genome-wide association study (GWAS) based on the genomes of 68 S. suis isolates. Through GWAS, a total number of 172 genes were identified. Among these genes, 143 of them distribute in virulent isolates. Further VFs interaction network analysis based on protein-protein interaction database found that 71 genes identified in this study could interact with known VFs and some of them even played an important role as the bridge between known VFs or formed important hub. In addition, 12 genes were found conserved in virulent isolates and 3 genes were conserved in avirulent isolates, 8 genes of the virulent conserved genes were belonging to a srtBCD pili cluster. Considering that sbp2', a member of the srtBCD pili cluster has been reported as a virulence-associated factor, we predict that sbp2' could be a fitness virulence-associated marker of virulent isolates. Taken together, our findings contribute to the insights in S. suis pathogenesis, enhance the knowledge of the genomic evolution of S. suis and provide several novel virulence-associated candidates.
Collapse
Affiliation(s)
- Genglin Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Xuewei Kong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Dechao Du
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Dan Wei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Yanfei Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Laboratory of Veterinary Biological Engineering and Technology of Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| |
Collapse
|
9
|
Lauer AN, Scholtysik R, Beineke A, Baums CG, Klose K, Valentin-Weigand P, Ishikawa H, Schroten H, Klein-Hitpass L, Schwerk C. A Comparative Transcriptome Analysis of Human and Porcine Choroid Plexus Cells in Response to Streptococcus suis Serotype 2 Infection Points to a Role of Hypoxia. Front Cell Infect Microbiol 2021; 11:639620. [PMID: 33763387 PMCID: PMC7982935 DOI: 10.3389/fcimb.2021.639620] [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: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 11/14/2022] Open
Abstract
Streptococcus suis (S. suis) is an important opportunistic pathogen, which can cause septicemia and meningitis in pigs and humans. Previous in vivo observations in S. suis-infected pigs revealed lesions at the choroid plexus (CP). In vitro experiments with primary porcine CP epithelial cells (PCPEC) and human CP epithelial papilloma (HIBCPP) cells demonstrated that S. suis can invade and traverse the CP epithelium, and that the CP contributes to the inflammatory response via cytokine expression. Here, next generation sequencing (RNA-seq) was used to compare global transcriptome profiles of PCPEC and HIBCPP cells challenged with S. suis serotype (ST) 2 infected in vitro, and of pigs infected in vivo. Identified differentially expressed genes (DEGs) were, amongst others, involved in inflammatory responses and hypoxia. The RNA-seq data were validated via quantitative PCR of selected DEGs. Employing Gene Set Enrichment Analysis (GSEA), 18, 28, and 21 enriched hallmark gene sets (GSs) were identified for infected HIBCPP cells, PCPEC, and in the CP of pigs suffering from S. suis ST2 meningitis, respectively, of which eight GSs overlapped between the three different sample sets. The majority of these GSs are involved in cellular signaling and pathways, immune response, and development, including inflammatory response and hypoxia. In contrast, suppressed GSs observed during in vitro and in vivo S. suis ST2 infections included those, which were involved in cellular proliferation and metabolic processes. This study suggests that similar cellular processes occur in infected human and porcine CP epithelial cells, especially in terms of inflammatory response.
Collapse
Affiliation(s)
- Alexa N Lauer
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rene Scholtysik
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| | - Andreas Beineke
- Institute for Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Christoph Georg Baums
- Faculty of Veterinary Medicine, Institute of Bacteriology and Mycology, Leipzig University, Leipzig, Germany
| | - Kristin Klose
- Faculty of Veterinary Medicine, Institute of Veterinary Pathology, Leipzig University, Leipzig, Germany
| | | | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
10
|
Ma J, Zhang Z, Pan Z, Bai Q, Zhong X, Zhu Y, Zhang Y, Wu Z, Liu G, Yao H. Streptococcus suis Uptakes Carbohydrate Source from Host Glycoproteins by N-glycans Degradation System for Optimal Survival and Full Virulence during Infection. Pathogens 2020; 9:E387. [PMID: 32443590 PMCID: PMC7281376 DOI: 10.3390/pathogens9050387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
Infection with the epidemic virulent strain of Streptococcus suis serotype 2 (SS2) can cause septicemia in swine and humans, leading to pneumonia, meningitis and even cytokine storm of Streptococcal toxic shock-like syndrome. Despite some progress concerning the contribution of bacterial adhesion, biofilm, toxicity and stress response to the SS2 systemic infection, the precise mechanism underlying bacterial survival and growth within the host bloodstream remains elusive. Here, we reported the SS2 virulent strains with a more than 20 kb endoSS-related insertion region that showed significantly higher proliferative ability in swine serum than low-virulent strains. Further study identified a complete N-glycans degradation system encoded within this insertion region, and found that both GH92 and EndoSS contribute to bacterial virulence, but that only DndoSS was required for optimal growth of SS2 in host serum. The supplement of hydrolyzed high-mannose-containing glycoprotein by GH92 and EndoSS could completely restore the growth deficiency of endoSS deletion mutant in swine serum. EndoSS only hydrolyzed a part of the model glycoprotein RNase B with high-mannose N-linked glycoforms into a low molecular weight form, and the solo activity of GH92 could not show any changes comparing with the blank control in SDS-PAGE gel. However, complete hydrolyzation was observed under the co-incubation of EndoSS and GH92, suggesting GH92 may degrade the high-mannose arms of N-glycans to generate a substrate for EndoSS. In summary, these findings provide compelling evidences that EndoSS-related N-glycans degradation system may enable SS2 to adapt to host serum-specific availability of carbon sources from glycoforms, and be required for optimal colonization and full virulence during systemic infection.
Collapse
Affiliation(s)
- Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Ze Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Zihao Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Qiankun Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Xiaojun Zhong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Yinchu Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Yue Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Guangjin Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (J.M.); (Z.Z.); (Z.P.); (Q.B.); (X.Z.); (Y.Z.); (Y.Z.); (Z.W.); (G.L.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| |
Collapse
|
11
|
Song H, Kang Y, Qian A, Shan X, Li Y, Zhang L, Zhang H, Sun W. Inactivation of the T6SS inner membrane protein DotU results in severe attenuation and decreased pathogenicity of Aeromonas veronii TH0426. BMC Microbiol 2020; 20:76. [PMID: 32245412 PMCID: PMC7119292 DOI: 10.1186/s12866-020-01743-5] [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: 12/27/2019] [Accepted: 03/04/2020] [Indexed: 11/19/2022] Open
Abstract
Background The inner membrane protein DotU of Aeromonas veronii is an important component of the minimal core conserved membrane proteome required for the formation of an envelope-transmembrane complex. This protein functions in a type VI secretion system (T6SS), and the role of this T6SS during the pathogenic process has not been clearly described. Results A recombinant A. veronii with a partial disruption of the dotU gene (720 bp of the in-frame sequence) (defined as ∆dotU) was constructed by two conjugate exchanges. We found that the mutant ∆dotU allele can be stably inherited for more than 50 generations. Inactivation of the A. veronii dotU gene resulted in no significant changes in growth or resistance to various environmental changes. However, compared with the wild-type strain colony, the mutant ∆dotU colony had a rough surface morphology. In addition, the biofilm formation ability of the mutant ∆dotU was significantly enhanced by 2.1-fold. Conversely, the deletion of the dotU gene resulted in a significant decrease in pathogenicity and infectivity compared to those of the A. veronii wild-type strain. Conclusions Our findings indicated that the dotU gene was an essential participant in the pathogenicity and invasiveness of A. veronii TH0426, which provides a novel perspective on the pathogenesis of TH0426 and lays the foundation for discovering potential T6SS effectors.
Collapse
Affiliation(s)
- Haichao Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Yuanhuan Kang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Aidong Qian
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Xiaofeng Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Ying Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Lei Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Haipeng Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Wuwen Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, China.
| |
Collapse
|
12
|
Si Y, Zhang L, Zhang L, Zhao F, Wang Q, Qian G, Yin S. Transcriptome analysis provides insight into the role of the melanin pathway in two differently pigmented strains of the turtle Pelodiscus sinensis. Dev Genes Evol 2019; 229:10.1007/s00427-019-00639-3. [PMID: 31712893 DOI: 10.1007/s00427-019-00639-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/16/2019] [Indexed: 11/29/2022]
Abstract
Pelodiscus sinensis is the most common turtle species raised in East and Southeast Asia. The Japanese strain and the Qingxi black strain are important aquatic breeds of P. sinensis in China with unique characteristics in terms of production performance and appearance, including skin color. In this study, melanin content measure and histological analysis on skin samples of these two strains were carried out to compare their color characteristics. The results showed that Qingxi black turtles clearly have a greater ability to deposit melanin than the Japanese strain especially in the abdomen. Then, de novo transcriptome assembly and differential expression profiling analyses on the ventral skin from the two strains were performed to identify the genes responsible for the differences in skin color using the Illumina RNA-Seq system with three biological replicates. A total of 19,331 annotated unigenes were found by aligning to the reference genome of P. sinensis using TopHat v2.0.12. Differential expression analysis revealed that 670 genes were expressed differently, including 185 upregulated genes and 485 downregulated genes in Qingxi black strain using the DESeq R package (|log2FoldChange| ≥ 1, padj < 0.05). Sixteen differentially expressed genes (DEGs), which were randomly selected, were confirmed by quantitative real-time PCR (qRT-PCR). GO and KEGG analyses revealed four DEGs (agouti signaling protein, frizzled family receptor 1, phospholipase C, and protein kinase C) were related to melanogenesis pathway. Gene expression levels of the four DEGs as well as three genes from the tyrosinase gene family were measured by qRT-PCR. The results indicated that agouti signaling protein, tyrosinase-related protein, and dopachrome tautomerase could be the main genes responsible for the difference in abdominal skin color between the two turtle strains. This study provided valuable information for further analysis of the melanogenesis mechanisms in different varieties of P. sinensis.
Collapse
Affiliation(s)
- Yuexiu Si
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China.
| | - Lili Zhang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, People's Republic of China
| | - Linmeng Zhang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, People's Republic of China
| | - Feng Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Qian Wang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, People's Republic of China
| | - Guoying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, People's Republic of China
| | - Shangjun Yin
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China.
| |
Collapse
|
13
|
Du F, Lv X, Duan D, Wang L, Huang J. Characterization of a Linezolid- and Vancomycin-Resistant Streptococcus suis Isolate That Harbors optrA and vanG Operons. Front Microbiol 2019; 10:2026. [PMID: 31551963 PMCID: PMC6746840 DOI: 10.3389/fmicb.2019.02026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Linezolid and vancomycin are among the last-resort antimicrobial agents in the treatment of multidrug-resistant Gram-positive bacterial infections. Linezolid- and vancomycin-resistant (LVR) Gram-positive bacteria may pose severe threats to public health. In this study, three optrA- and vanG-positive Streptococcus suis strains were isolated from two farms of different cities. There were only 1 and 343 single-nucleotide polymorphisms in coding region (cSNPs) of HCB4 and YSJ7 to YSJ17, respectively. Mobilome analysis revealed the presence of vanG, erm(B), tet(O/W/32/O), and aadE-apt-sat4-aphA3 cluster on an integrative and conjugative element, ICESsuYSJ17, and erm(B), aphA3, aac(6')-aph(2″), catpC194, and optrA on a prophage, ΦSsuYSJ17-3. ICESsuYSJ17 exhibited a mosaic structure and belongs to a highly prevalent and transferable ICESa2603 family of Streptococcus species. ΦSsuYSJ17-3 shared conserved backbone to a transferable prophage Φm46.1. A novel composite transposon, IS1216E-araC-optrA-hp-catpC194-IS1216E, which can be circulated as translocatable unit (TU) by IS1216E, was integrated on ΦSsuYSJ17-3. Vancomycin resistance phenotype and vanG transcription assays revealed that the vanG operon was inducible. The LVR strain YSJ17 exhibited moderate virulence in a zebrafish infection model. To our knowledge, this is the first report of LVR isolate, which is mediated by acquired resistance genes optrA and vanG operons in Gram-positive bacteria. Since S. suis has been recognized as an antimicrobial resistance reservoir in the spread of resistance genes to major streptococcal pathogens, the potential risks of disseminating of optrA and vanG from S. suis to other Streptococcus spp. are worrisome and routine surveillance should be strengthened.
Collapse
Affiliation(s)
- Fanshu Du
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xi Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Duan Duan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
14
|
Huang J, Sun J, Wu Y, Chen L, Duan D, Lv X, Wang L. Identification and pathogenicity of an XDR Streptococcus suis isolate that harbours the phenicol-oxazolidinone resistance genes optrA and cfr, and the bacitracin resistance locus bcrABDR. Int J Antimicrob Agents 2019; 54:43-48. [PMID: 30981924 DOI: 10.1016/j.ijantimicag.2019.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/30/2022]
Abstract
One hundred and seven Streptococcus suis isolates were collected from healthy pigs or asymptomatic carriers in Jiangsu, China in 2016-2017. Thirty-eight percent of the isolates were linezolid-resistant and all carried the optrA gene. Among them, one isolate, SFJ44, was resistant to all 20 of the antibiotics tested, except for ceftiofur, and thus exhibited an extensively-drug-resistant phenotype. This isolate carried the optrA gene and the bacitracin resistance locus bcrABDR on an antibiotic-resistance-associated genomic island (ARGI1), and harboured the resistance genes cfr, aadE, sat4, spw-like, aphA3, mef(A), msr(D), erm(A)-like, erm(B), tetAB(P)', tet(M) and catQ on ARGI2∼4. The IS1216E-bcrABDR-ISEnfa1 segment showed >99.9% sequence identity to corresponding sequences from other species. The cfr gene was located on ARGI4, and two IS6 family insertion sequences, IS1216E and ISTeha2, were found upstream and downstream of cfr-ΔISEnfa5, respectively. A circular intermediate of bcrABDR-ISEnfa1 was detected, suggesting the role of ISEnfa1 in dissemination of bcrABDR. Other antibiotic resistance genes might be acquired from different Gram-positive pathogens. Infection of zebrafish showed that SFJ44 exhibited a virulence level comparable to serotype 2 hypervirulent strain SC070731, highlighting the need for surveillance of the pathogenicity of multi-drug-resistant S. suis isolates. This is the first report of the co-existence of optrA and cfr, and of the bcrABDR locus in streptococci. As it has been suggested that S. suis may act as an antibiotic resistance reservoir contributing to the spread of resistance genes to major streptococcal pathogens, the potential dissemination of these resistance genes among Gram-positive bacteria is of concern and routine surveillance should be strengthened.
Collapse
Affiliation(s)
- Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Junjie Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuanchang Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Li Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Duan Duan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xi Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
15
|
Huang J, Liu X, Chen H, Chen L, Gao X, Pan Z, Wang J, Lu C, Yao H, Wang L, Wu Z. Identification of six novel capsular polysaccharide loci (
NCL
) from
Streptococcus suis
multidrug resistant non‐typeable strains and the pathogenic characteristic of strains carrying new
NCL
s. Transbound Emerg Dis 2019; 66:995-1003. [DOI: 10.1111/tbed.13123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/06/2019] [Accepted: 01/09/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
| | - Xi Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Hao Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Li Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
| | - Xueping Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Zihao Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Jian Wang
- Shanghai Municipal Animal Disease Control Center Shanghai China
| | - Chengping Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety College of Veterinary Medicine Nanjing Agricultural University Nanjing China
- Key Lab of Animal Bacteriology, Ministry of Agriculture Nanjing China
- OIE Reference Lab for Swine Streptococcosis Nanjing China
| |
Collapse
|
16
|
Alves‐Barroco C, Roma‐Rodrigues C, Raposo LR, Brás C, Diniz M, Caço J, Costa PM, Santos‐Sanches I, Fernandes AR. Streptococcus dysgalactiae subsp. dysgalactiae isolated from milk of the bovine udder as emerging pathogens: In vitro and in vivo infection of human cells and zebrafish as biological models. Microbiologyopen 2019; 8:e00623. [PMID: 29577680 PMCID: PMC6341033 DOI: 10.1002/mbo3.623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 12/23/2022] Open
Abstract
Streptococcus dysgalactiae subsp. dysgalactiae (SDSD) is a major cause of bovine mastitis and has been regarded as an animal-restricted pathogen, although rare infections have been described in humans. Previous studies revealed the presence of virulence genes encoded by phages of the human pathogen Group A Streptococcus pyogenes (GAS) in SDSD isolated from the milk of bovine udder with mastitis. The isolates SDSD VSD5 and VSD13 could adhere and internalize human primary keratinocyte cells, suggesting a possible human infection potential of bovine isolates. In this work, the in vitro and in vivo potential of SDSD to internalize/adhere human cells of the respiratory track and zebrafish as biological models was evaluated. Our results showed that, in vitro, bovine SDSD strains could interact and internalize human respiratory cell lines and that this internalization was dependent on an active transport mechanism and that, in vivo, SDSD are able to cause invasive infections producing zebrafish morbidity and mortality. The infectious potential of these isolates showed to be isolate-specific and appeared to be independent of the presence or absence of GAS phage-encoded virulence genes. Although the infection ability of the bovine SDSD strains was not as strong as the human pathogenic S. pyogenes in the zebrafish model, results suggested that these SDSD isolates are able to interact with human cells and infect zebrafish, a vertebrate infectious model, emerging as pathogens with zoonotic capability.
Collapse
Affiliation(s)
- Cinthia Alves‐Barroco
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Catarina Roma‐Rodrigues
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Luís R. Raposo
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Catarina Brás
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Mário Diniz
- Departamento de QuímicaFaculdade de Ciências e TecnologiaUCIBIOUniversidade NOVA de LisboaCaparicaPortugal
| | - João Caço
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Pedro M. Costa
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
- MARE ‐ Marine and Environmental Sciences CentreDepartamento de Ciências e Engenharia do AmbienteFaculdade de Ciências e TecnologiaUniversidade Nova de LisboaCaparicaPortugal
| | - Ilda Santos‐Sanches
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| | - Alexandra R. Fernandes
- Departamento de Ciências da VidaFaculdade de Ciências e TecnologiaUCIBIOUniversidade Nova de LisboaCaparicaPortugal
| |
Collapse
|
17
|
Streptococcus suis biofilm: regulation, drug-resistance mechanisms, and disinfection strategies. Appl Microbiol Biotechnol 2018; 102:9121-9129. [PMID: 30209548 DOI: 10.1007/s00253-018-9356-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/30/2018] [Indexed: 10/28/2022]
Abstract
Streptococcus suis (S. suis) is a major swine pathogen and an important zoonotic agent. Like most pathogens, the ability of S. suis to form biofilms plays a significant role in its virulence and drug resistance. A better understanding of the mechanisms involved in biofilm formation by S. suis as well as of the methods to efficiently remove and kill biofilm-embedded bacteria can be of high interest for the prevention and treatment of S. suis infections. The aim of this literature review is to update our current knowledge of S. suis biofilm formation, regulatory mechanisms, drug-resistance mechanisms, and disinfection strategies.
Collapse
|
18
|
Zhang Q, Ji C, Ren J, Zhang Q, Dong X, Zu Y, Jia L, Li W. Differential transcriptome analysis of zebrafish (Danio rerio) larvae challenged by Vibrio parahaemolyticus. JOURNAL OF FISH DISEASES 2018; 41:1049-1062. [PMID: 29572872 DOI: 10.1111/jfd.12796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 06/08/2023]
Abstract
Zebrafish embryo and larva represent a useful in vivo model for identification of host innate immune responses to bacterial infection. Vibrio parahaemolyticus is a typical zoonotic pathogen worldwide that causes acute gastroenteritis in humans and vibriosis in fishes. However, the mechanism of the innate immune response in the zebrafish larvae infected by V. parahaemolyticus has not been clear. We analysed the transcriptomic profile of 3 days post-fertilization (dpf) zebrafish larvae immersed in V. parahaemolyticus 13 (Vp13) strain suspension for 2 hr. A total of 602 differentially expressed genes (DEGs) were identified in the infection group, of which 175 (29.07%) genes were upregulated and 427 (70.93%) genes were downregulated. These altered genes encoded complement and coagulation cascades, chemokine, TNF signalling pathway, NF-κB signalling pathway and JAK-STAT signalling pathway. Some significant DEGs, such as mmp13, cxcr4a, ccl20, hsp70, gngt, serpina1l, il8, cofilin and il11, were subjected to quantitative gene expression analysis, and the results were consistent with those of the transcriptome profile. These results clearly demonstrated that exposure to V. parahaemolyticus for 2 hr could activate innate immune response in 3dpf larvae by altered expression of downstream signalling pathway genes of pattern recognition receptors (PRRs). Our results also provide a useful reference for future analysis of signal transduction pathways and pathogenesis mechanisms underlying the systemic innate immune response to the external bacteria of V. parahaemolyticus.
Collapse
Affiliation(s)
- Qinghua Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Ce Ji
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Jianfeng Ren
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Qiuyue Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Xuehong Dong
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Yao Zu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Liang Jia
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Weiming Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
19
|
Leigh WJ, Zadoks RN, Jaglarz A, Costa JZ, Foster G, Thompson KD. Evaluation of PCR primers targeting the groEL gene for the specific detection of Streptococcus agalactiae in the context of aquaculture. J Appl Microbiol 2018; 125:666-674. [PMID: 29786935 DOI: 10.1111/jam.13925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 02/06/2023]
Abstract
AIMS The aim of this study was to design a set of primers for specific detection and identification of Streptococcus agalactiae in polymerase chain reaction (PCR) that can detect a diverse range of S. agalactiae isolates from different hosts and that it is capable of discriminating between S. agalactiae and other species that are closely related or potentially present in aquaculture environments, notably Streptococcus iniae. METHODS AND RESULTS Primers, based on the groEL2 gene of S. agalactiae, were shown to be epidemiologically sensitive to 97 isolates of S. agalactiae, representing 11 clonal complexes derived from piscine, terrestrial and aquatic mammalian host species. The primers were tested with 10 S. iniae isolates and 22 other comparator species with no cross-reaction observed after optimization of reaction conditions. They have a high analytical sensitivity, detecting as few as 10 copies of S. agalactiae genomic DNA per reaction and are capable of detecting the target in DNA extracted from the brains of infected fish. CONCLUSIONS The primers proved suitable for the sensitive and specific detection of S. agalactiae from dairy-, human- and fish-related origins by PCR. SIGNIFICANCE AND IMPACT OF THE STUDY Due to the importance of S. agalactiae as a pathogen, many PCR primers have been published for this bacterium, designed largely for its detection in dairy and human samples, but many cross-reacting with S. iniae. The ability to differentiate between S. agalactiae and S. iniae in aquaculture derived samples is important as both infect fish, causing similar disease symptoms and are phenotypically similar, yet control strategies and zoonotic risk are species specific.
Collapse
Affiliation(s)
- W J Leigh
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK.,Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - R N Zadoks
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK.,Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - A Jaglarz
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK
| | - J Z Costa
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK
| | - G Foster
- SAC Consulting Veterinary Services, Drummond Hill, Stratherrick Road, Inverness, UK
| | - K D Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik, UK
| |
Collapse
|
20
|
The Two-Component Signaling System VraSR ss Is Critical for Multidrug Resistance and Full Virulence in Streptococcus suis Serotype 2. Infect Immun 2018; 86:IAI.00096-18. [PMID: 29685990 DOI: 10.1128/iai.00096-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/18/2018] [Indexed: 11/20/2022] Open
Abstract
Streptococcus suis has received increasing attention for its involvement in severe human infections worldwide as well as in multidrug resistance. Two-component signaling systems (TCSSs) play important roles in bacterial adaptation to various environmental stimuli. In this study, we identified a novel TCSS located in S. suis serotype 2 (SS2), designated VraSRSS, which is involved in bacterial pathogenicity and susceptibility to antimicrobials. Our data demonstrated that the yvqFSS gene, located upstream of vraSRSS , shared the same promoter with the TCSS genes, which was directly regulated by VraSRSS, as shown in electrophoretic mobility shift assays. Notably, YvqFSS and VraSRSS constitute a novel multidrug resistance module of SS2 that participates in resistance to certain groups of antimicrobials. Further analyses showed that VraSRSS inactivation significantly attenuated bacterial virulence in animal models, which, coupled with the significant activation of VraSRSS expression observed in host blood, strongly suggested that VraSRSS is an important regulator of SS2 pathogenicity. Indeed, RNA-sequencing analyses identified 106 genes that were differentially expressed between the wild-type and ΔvraSRSS strains, including genes involved in capsular polysaccharide (CPS) biosynthesis. Subsequent studies confirmed that VraSRSS indirectly regulated the transcription of CPS gene clusters and, thus, controlled the CPS thickness shown by transmission electron microscopy. Decreased CPS biosynthesis caused by vraSRSS deletion subsequently increased bacterial adhesion to epithelial cells and attenuated antiphagocytosis against macrophages, which partially clarified the pathogenic mechanism mediated by VraSRSS Taken together, our data suggest that the novel TCSS, VraSRSS, plays critical roles for multidrug resistance and full virulence in SS2.
Collapse
|
21
|
Li Q, Fu Y, He Y, Zhang Y, Qian Y, Yu Y, Yao H, Lu C, Zhang W. Fibronectin-/fibrinogen-binding protein (FBPS) is not a critical virulence factor for the Streptococcus suis serotype 2 strain ZY05719. Vet Microbiol 2017; 208:38-46. [DOI: 10.1016/j.vetmic.2017.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/09/2017] [Accepted: 07/09/2017] [Indexed: 01/06/2023]
|
22
|
Critical Streptococcus suis Virulence Factors: Are They All Really Critical? Trends Microbiol 2017; 25:585-599. [PMID: 28274524 DOI: 10.1016/j.tim.2017.02.005] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/30/2017] [Accepted: 02/09/2017] [Indexed: 01/15/2023]
Abstract
Streptococcus suis is an important swine pathogen that can be transmitted to humans by contact with diseased animals or contaminated raw pork products. This pathogen possesses a coat of capsular polysaccharide (CPS) that confers protection against the immune system. Yet, the CPS is not the only virulence factor enabling this bacterium to successfully colonize, invade, and disseminate in its host leading to severe systemic diseases such as meningitis and toxic shock-like syndrome. Indeed, recent research developments, cautiously inventoried in this review, have revealed over 100 'putative virulence factors or traits' (surface-associated or secreted components, regulatory genes or metabolic pathways), of which at least 37 have been claimed as being 'critical' for virulence. In this review we discuss the current contradictions and controversies raised by this explosion of virulence factors and the future directions that may be conceived to advance and enlighten research on S. suis pathogenesis.
Collapse
|
23
|
Xiao G, Wu Z, Zhang S, Tang H, Wang F, Lu C. Mac Protein is not an Essential Virulence Factor for the Virulent Reference Strain Streptococcus suis P1/7. Curr Microbiol 2016; 74:90-96. [PMID: 27847975 DOI: 10.1007/s00284-016-1160-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 11/08/2016] [Indexed: 11/25/2022]
Abstract
Streptococcus suis is a major pathogen of pigs and also an important zoonotic agent for humans. A S. suis protein containing Mac-1 domain (designated Mac) is a protective antigen, exclusively cleaves porcine IgM, and contributes to complement evasion with the presence of high titers of specific porcine anti-S. suis IgM, but its role in S. suis virulence has not been investigated in natural healthy host without specific IgM. In this study, a mac deletion mutant was constructed by homologous recombination in S. suis serotype 2 virulent reference strain P1/7. Deletion of mac did not significantly influence phagocytosis or intracellular survival within murine macrophages RAW264.7, or the oxidative-burst induction of RAW264.7 and murine neutrophils. Furthermore, the mutant is as virulent as the wild-type strain in pig, mouse, and zebrafish infection models. Our data suggest that Mac is not essential for S. suis virulence in strain P1/7 in natural healthy host without specific IgM, and the immunogenicity of Mac does not appear to correlate with its significance for virulence.
Collapse
Affiliation(s)
- Genhui Xiao
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Zongfu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Shouming Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Huanyu Tang
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Fengqiu Wang
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China
| | - Chengping Lu
- College of Veterinary Medicine, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing, 210095, China.
| |
Collapse
|
24
|
Xu J, Zhao J, Li Y, Zou Y, Lu B, Chen Y, Ma Y, Xu H. Evaluation of differentially expressed immune-related genes in intestine of Pelodiscus sinensis after intragastric challenge with lipopolysaccharide based on transcriptome analysis. FISH & SHELLFISH IMMUNOLOGY 2016; 56:417-426. [PMID: 27475104 DOI: 10.1016/j.fsi.2016.07.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/01/2016] [Accepted: 07/25/2016] [Indexed: 06/06/2023]
Abstract
Pelodiscus sinensis is the most common turtle species that has been raised in East and Southeast Asia. However, there are still limited studies about the immune defense mechanisms in its small intestine until now. In the present research, histological analysis and transcriptome analysis was performed on the small intestine of P. sinensis after intragastric challenge with LPS to explore its mechanisms of immune responses to pathogens. The result showed the number of intraepithelial lymphocytes (IELs) and goblet cells (GCs) in its intestine increased significantly at 48 h post-challenge with LPS by intragastrical route, indicating clearly the intestinal immune response was induced. Compared with the control, a total of 748 differentially expressed genes (DEGs) were identified, including 361 up-regulated genes and 387 down-regulated genes. Based on the Gene Ontology (GO) annotation and the Kyoto Encyclopedia of Genes and Genomes (KEGG), 48 immune-related DEGs were identified, which were classified into 82 GO terms and 14 pathways. Finally, 18 DEGs, which were randomly selected, were confirmed by quantitative real-time PCR (qRT-PCR). Our results provide valuable information for further analysis of the immune defense mechanisms against pathogens in the small intestine of P. sinensis.
Collapse
Affiliation(s)
- Jiehao Xu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Jing Zhao
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Yiqun Li
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Yiyi Zou
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Binjie Lu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Yuyin Chen
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Youzhi Ma
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China
| | - Haisheng Xu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, China.
| |
Collapse
|
25
|
Shan Y, Zhang Y, Zhuo X, Li X, Peng J, Fang W. Matrix metalloproteinase-9 plays a role in protecting zebrafish from lethal infection with Listeria monocytogenes by enhancing macrophage migration. FISH & SHELLFISH IMMUNOLOGY 2016; 54:179-187. [PMID: 27068748 DOI: 10.1016/j.fsi.2016.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/19/2016] [Accepted: 04/07/2016] [Indexed: 06/05/2023]
Abstract
Zebrafish could serve as an alternative animal model for pathogenic bacteria in multiple infectious routes. Our previous study showed that immersion infection in zebrafish with Listeria monocytogenes did not cause lethality but induced transient expression of several immune response genes. We used an Affymetrix gene chip to examine the expression profiles of genes of zebrafish immersion-infected with L. monocytogenes. A total of 239 genes were up-regulated and 56 genes down-regulated compared with uninfected fish. Highest expression (>20-fold) was seen with the mmp-9 gene encoding the matrix metalloproteinase-9 (Mmp-9) known to degrade the extracellular matrix proteins. By morpholino knockdown of mmp-9, we found that the morphants showed rapid death with much higher bacterial load after intravenous or intraventricular (brain ventricle) infection with L. monocytogenes. Macrophages in mmp-9-knockdown morphants had significant defect in migrating to the brain cavity upon intraventricular infection. Decreased migration of murine macrophages with knockdown of mmp-9 and cd44 was also seen in transwell inserts with 8-μm pore polycarbonate membrane, as compared with the scrambled RNA. These findings suggest that Mmp-9 is a protective molecule against infection by L. monocytogenes by engaging in migration of zebrafish macrophages to the site of infection via a non-proteolytic role. Further work is required on the molecular mechanisms governing Mmp-9-driven macrophage migration in zebrafish.
Collapse
Affiliation(s)
- Ying Shan
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA
| | - Yikai Zhang
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA
| | - Xunhui Zhuo
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA
| | - Xiaoliang Li
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA
| | - Jinrong Peng
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA
| | - Weihuan Fang
- Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, CHINA.
| |
Collapse
|
26
|
Wang B, Gan Z, Cai S, Wang Z, Yu D, Lin Z, Lu Y, Wu Z, Jian J. Comprehensive identification and profiling of Nile tilapia (Oreochromis niloticus) microRNAs response to Streptococcus agalactiae infection through high-throughput sequencing. FISH & SHELLFISH IMMUNOLOGY 2016; 54:93-106. [PMID: 27050313 DOI: 10.1016/j.fsi.2016.03.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/22/2016] [Accepted: 03/26/2016] [Indexed: 06/05/2023]
Abstract
MicroRNAs are a kind of small non-coding RNAs that participate in various biological processes. Deregulated microRNA expression is associated with several types of diseases. Tilapia (Oreochromis niloticus) is an important commercial fish species in China. To identify miRNAs and investigate immune-related miRNAs of O. niloticus, we applied high-throughput sequencing technology to identify and analyze miRNAs from tilapia infected with Streptococcus agalactiae at a timescale of 72 h divided into six different time points. The results showed that a total of 3009 tilapia miRNAs were identified, including in 1121 miRNAs which have homologues in the currently available databases and 1878 novel miRNAs. The expression levels of 218 tilapia miRNAs were significantly altered at 6 h-72 h post-bacterial infection (pi), and these miRNAs were therefore classified as differentially expressed tilapia miRNAs. For the 1121 differentially expressed tilapia miRNAs target 41961 genes. GO and KEGG enrichment analysis revealed that some target genes of tilapia miRNAs were grouped mainly into the categories of apoptotic process, signal pathway, and immune response. This is the first report of comprehensive identification of O. niloticus miRNAs being differentially regulated in spleen in normal conditions relating to S. agalactiae infection. This work provides an opportunity for further understanding of the molecular mechanisms of miRNA regulation in O. niloticus host-pathogen interactions.
Collapse
Affiliation(s)
- Bei Wang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Zhen Gan
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Shuanghu Cai
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Zhongliang Wang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Dapeng Yu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Ziwei Lin
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Zaohe Wu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animala, Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China.
| |
Collapse
|
27
|
Xu C, Evensen Ø, Munang'andu HM. A de novo transcriptome analysis shows that modulation of the JAK-STAT signaling pathway by salmonid alphavirus subtype 3 favors virus replication in macrophage/dendritic-like TO-cells. BMC Genomics 2016; 17:390. [PMID: 27215196 PMCID: PMC4878077 DOI: 10.1186/s12864-016-2739-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 05/12/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The Janus kinase (Jak) and signaling transducer activator of transcription (Stat) pathway mediates the signaling of genes required for cellular development and homeostasis. To elucidate the effect of type I IFN on the Jak/stat pathway in salmonid alphavirus subtype 3 (SAV3) infected macrophage/dendritic like TO-cells derived from Atlantic salmon (Salmo salar L) headkidney leukocytes, we used a differential transcriptome analysis by RNA-seq and the Kyoto encyclopedia of genes and genomes (KEGGs) pathway analysis to generate a repertoire of de novo assembled genes from type I IFN treated and non-treated TO-cells infected with SAV3. RESULTS Concurrent SAV3 infection with type I IFN treatment of TO-cells suppressed SAV3 structural protein (SP) expression by 2log10 at 2 days post infection compared to SAV3 infection without IFN treatment which paved way to evaluating the impact of type I IFN on expression of Jak/stat pathway genes in SAV3 infected TO-cells. In the absence of type I IFN treatment, SAV3 downregulated several Jak/stat pathway genes that included type I and II receptor genes, Jak2, tyrosine kinase 2 (Tyk2), Stat3 and Stat5 pointing to possible failure to activate the Jak/stat signaling pathway and inhibition of signal transducers caused by SAV3 infection. Although the suppressor of cytokine signaling (SOCS) genes 1 and 3 were upregulated in the IFN treated cells, only SOCS3 was downregulated in the SAV3 infected cells which points to inhibition of SOCS3 by SAV3 infection in TO-cells. CONCLUSION Data presented in this study shows that SAV3 infection downregulates several genes of the Jak/stat pathway, which could be an immune evasion strategy, used to block the transcription of antiviral genes that would interfere with SAV3 replication in TO-cells. Overall, we have shown that combining de novo assembly with pathway based transcriptome analyses provides a contextual approach to understanding the molecular networks of genes that form the Jak/stat pathway in TO-cells infected by SAV3.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, P.O Box 8146, Oslo, NO-0033 Dep, Norway
| | - Øystein Evensen
- Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, P.O Box 8146, Oslo, NO-0033 Dep, Norway
| | - Hetron Mweemba Munang'andu
- Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, P.O Box 8146, Oslo, NO-0033 Dep, Norway.
| |
Collapse
|
28
|
Zaccaria E, Cao R, Wells JM, van Baarlen P. A Zebrafish Larval Model to Assess Virulence of Porcine Streptococcus suis Strains. PLoS One 2016; 11:e0151623. [PMID: 26999052 PMCID: PMC4801416 DOI: 10.1371/journal.pone.0151623] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/29/2016] [Indexed: 12/14/2022] Open
Abstract
Streptococcus suis is an encapsulated Gram-positive bacterium, and the leading cause of sepsis and meningitis in young pigs resulting in considerable economic losses in the porcine industry. It is also considered an emerging zoonotic agent. In the environment, both avirulent and virulent strains occur in pigs, and virulent strains appear to cause disease in both humans and pigs. There is a need for a convenient, reliable and standardized animal model to assess S. suis virulence. A zebrafish (Danio rerio) larvae infection model has several advantages, including transparency of larvae, low cost, ease of use and exemption from ethical legislation up to 6 days post fertilization, but has not been previously established as a model for S. suis. Microinjection of different porcine strains of S. suis in zebrafish larvae resulted in highly reproducible dose- and strain-dependent larval death, strongly correlating with presence of the S. suis capsule and to the original virulence of the strain in pigs. Additionally we compared the virulence of the two-component system mutant of ciaRH, which is attenuated for virulence in both mice and pigs in vivo. Infection of larvae with the ΔciaRH strain resulted in significantly higher survival rate compared to infection with the S10 wild-type strain. Our data demonstrate that zebrafish larvae are a rapid and reliable model to assess the virulence of clinical porcine S. suis isolates.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host-Microbe Interactomics, Department of Animal Sciences, Wageningen University, Wageningen, the Netherlands
| | - Rui Cao
- Host-Microbe Interactomics, Department of Animal Sciences, Wageningen University, Wageningen, the Netherlands
| | - Jerry M. Wells
- Host-Microbe Interactomics, Department of Animal Sciences, Wageningen University, Wageningen, the Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics, Department of Animal Sciences, Wageningen University, Wageningen, the Netherlands
- * E-mail:
| |
Collapse
|
29
|
Xu J, Mu Y, Zhang Y, Dong W, Zhu Y, Ma J, Song W, Pan Z, Lu C, Yao H. Antibacterial effect of porcine PTX3 against Streptococcus suis type 2 infection. Microb Pathog 2015; 89:128-39. [DOI: 10.1016/j.micpath.2015.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 09/07/2015] [Accepted: 09/24/2015] [Indexed: 11/28/2022]
|
30
|
Harvie EA, Huttenlocher A. Neutrophils in host defense: new insights from zebrafish. J Leukoc Biol 2015; 98:523-37. [PMID: 25717145 PMCID: PMC4569048 DOI: 10.1189/jlb.4mr1114-524r] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are highly motile phagocytic cells that play a critical role in the immune response to infection. Zebrafish (Danio rerio) are increasingly used to study neutrophil function and host-pathogen interactions. The generation of transgenic zebrafish lines with fluorescently labeled leukocytes has made it possible to visualize the neutrophil response to infection in real time by use of optically transparent zebrafish larvae. In addition, the genetic tractability of zebrafish has allowed for the generation of models of inherited neutrophil disorders. In this review, we discuss several zebrafish models of infectious disease, both in the context of immunocompetent, as well as neutrophil-deficient hosts and how these models have shed light on neutrophil behavior during infection.
Collapse
Affiliation(s)
- Elizabeth A Harvie
- *Microbiology Doctoral Training Program, Departments of Medical Microbiology and Immunology and Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Anna Huttenlocher
- *Microbiology Doctoral Training Program, Departments of Medical Microbiology and Immunology and Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
31
|
Liu G, Zhang W, Liu Y, Yao H, Lu C, Xu P. Identification of a virulence-related surface protein XF in piscine Streptococcus agalactiae by pre-absorbed immunoproteomics. BMC Vet Res 2014; 10:259. [PMID: 25344337 PMCID: PMC4219122 DOI: 10.1186/s12917-014-0259-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/17/2014] [Indexed: 02/06/2023] Open
Abstract
Background Since 2009, large-scale Streptococcus agalactiae infections have broken out in cultured tilapia farms in China, resulting in considerable economic losses. Screening of the surface proteins is required to identify virulence factors or protective antigens involved in piscine S.agalactiae infections in tilapia. Pre-absorbed immunoproteomics method (PAIM) is a useful method previously established in our laboratory for identifying bacterial surface proteins. Results A serine-rich repeat protein family 1 (Srr-1), designated XF, was identified by PAIM in piscine S. agalactiae isolate GD201008-001. To investigate the role of XF in the pathogenesis of piscine S. agalactiae, an isogenic xf mutant strain (Δxf) and a complemented strain (CΔxf) were successfully constructed. The Δxf mutant and CΔxf showed no significant differences in growth characteristics and adherence to HEp-2 cells compared with the wild-type strain. However the 50% lethal dose of Δxf was increased (4-fold) compared with that of the parental strain in a zebrafish infection model. Conclusions The findings demonstrated that XF is a virulence-related, highly immunoreactive surface protein and is involved in the pathogenicity of S. agalactiae infections in fish. Electronic supplementary material The online version of this article (doi:10.1186/s12917-014-0259-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guangjin Liu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Wei Zhang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Yongjie Liu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Huochun Yao
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Chengping Lu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Weigang No.1, Nanjing, 210095, Jiangsu, China.
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| |
Collapse
|
32
|
Wu Z, Wu C, Shao J, Zhu Z, Wang W, Zhang W, Tang M, Pei N, Fan H, Li J, Yao H, Gu H, Xu X, Lu C. The Streptococcus suis transcriptional landscape reveals adaptation mechanisms in pig blood and cerebrospinal fluid. RNA (NEW YORK, N.Y.) 2014; 20:882-898. [PMID: 24759092 PMCID: PMC4024642 DOI: 10.1261/rna.041822.113] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 03/11/2014] [Indexed: 06/03/2023]
Abstract
Streptococcus suis (SS) is an important pathogen of pigs, and it is also recognized as a zoonotic agent for humans. SS infection may result in septicemia or meningitis in the host. However, little is known about genes that contribute to the virulence process and survival within host blood or cerebrospinal fluid (CSF). Small RNAs (sRNA) have emerged as key regulators of virulence in several bacteria, but they have not been investigated in SS. Here, using a differential RNA-sequencing approach and RNAs from SS strain P1/7 grown in rich medium, pig blood, or CSF, we present the SS genome-wide map of 793 transcriptional start sites and 370 operons. In addition to identifying 29 sRNAs, we show that five sRNA deletion mutants attenuate SS virulence in a zebrafish infection model. Homology searches revealed that 10 sRNAs were predicted to be present in other pathogenic Streptococcus species. Compared with wild-type strain P1/7, sRNAs rss03, rss05, and rss06 deletion mutants were significantly more sensitive to killing by pig blood. It is possible that rss06 contributes to SS virulence by indirectly activating expression of SSU0308, a virulence gene encoding a zinc-binding lipoprotein. In blood, genes involved in the synthesis of capsular polysaccharide (CPS) and subversion of host defenses were up-regulated. In contrast, in CSF, genes for CPS synthesis were down-regulated. Our study is the first analysis of SS sRNAs involved in virulence and has both improved our understanding of SS pathogenesis and increased the number of sRNAs known to play definitive roles in bacterial virulence.
Collapse
Affiliation(s)
- Zongfu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Jing Shao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Weixue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | | | - Min Tang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | - Na Pei
- BGI-Shenzhen, Shenzhen 518083, China
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | | | - Huochun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| | - Hongwei Gu
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Chengping Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China
- OIE Reference Laboratory for Swine Streptococcosis, Nanjing 210095, China
| |
Collapse
|
33
|
Zhang L, Sun C, Ye X, Zou S, Lu M, Liu Z, Tian Y. Characterization of four heat-shock protein genes from Nile tilapia (Oreochromis niloticus) and demonstration of the inducible transcriptional activity of Hsp70 promoter. FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:221-33. [PMID: 23912482 DOI: 10.1007/s10695-013-9838-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 07/26/2013] [Indexed: 05/26/2023]
Abstract
Heat-shock proteins (Hsps), known as stress proteins and extrinsic chaperones, play important roles in the folding, translocation, and refolding/degradation of proteins. In this study, we identified four Hsps in Nile tilapia (Oreochromis niloticus), which display conserved Hsp characteristics in their predicted amino acid sequences. Further analyses on the structures, homology, and phylogenetics revealed that the four Hsps belong to Hsp70 family. One of them does not contain introns and is named Hsp70, while all the other three contain introns and are named Hsc70-1, Hsc70-2, and Hsc70-3. Expressions of the four Hsp proteins were observed in all examined tissues. Six hours after infection of Streptococcus agalactiae in Nile tilapia, the expression of Hsp70 was significantly increased in the liver, head kidney, spleen and gill, while Hsc70s' expression was unchanged in all examined tissues except the head kidney that showed significantly reduced expression of both Hsc70-2 and Hsc70-3. These results suggest that Hsp70 may participate in the defense against S. agalactiae infection. We then isolated the promoter of Hsp70 gene and inserted it into the donor plasmid of Tgf2 transposon system containing green fluorescent protein (GFP) gene. The plasmid was microinjected into zebrafish embryos, where the expression of GFP was induced by heat shock, S. agalactiae immersion challenge, indicating that the isolated Hsp70 promoter has transcriptional activity and is inducible by both heat shock and bacterial challenge. This promoter may facilitate the future construction of disease-resistant transgenic fish. The work also contributes to the further study of immune response of tilapia after bacterial infection.
Collapse
Affiliation(s)
- Lili Zhang
- Key Laboratory of Tropical and Subtropical Fisheries Resource Application and Cultivation, China Ministry of Agriculture; Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Johnson AC, Turko AJ, Klaiman JM, Johnston EF, Gillis TE. Cold acclimation alters the connective tissue content of the zebrafish (Danio rerio) heart. J Exp Biol 2014; 217:1868-75. [DOI: 10.1242/jeb.101196] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Abstract
Thermal acclimation can alter cardiac function and morphology in a number of fish species, but little is known about the regulation of these changes. The purpose of the current study was to determine how cold acclimation affects zebrafish (Danio rerio) cardiac morphology, collagen composition, and connective tissue regulation. Heart volume, the thickness of the compact myocardium, collagen content, and collagen fiber composition were compared between control (27°C) and cold acclimated (20°C) zebrafish using serially sectioned hearts stained with picrosirius red. Collagen content and fiber composition of the pericardial membrane were also examined. Cold acclimation did not affect the volume of the contracted heart, however there was a significant decrease in the thickness of the compact myocardium. There was also a decrease in the collagen content of the compact myocardium and in amount of thick collagen fibers throughout the heart. Cold-acclimated zebrafish also increased expression of the gene transcript for matrix metalloproteinase 2, matrix metalloproteinase 9, tissue inhibitor of metalloproteinase 2, and collagen Type 1 α1. We propose that the reduction in the thickness of the compact myocardium as well as the change in collagen content may help to maintain the compliance of the ventricle as temperatures decrease. Together, these results clearly demonstrate that the zebrafish heart undergoes significant remodelling in response to cold acclimation.
Collapse
|
35
|
Wu Z, Wang W, Tang M, Shao J, Dai C, Zhang W, Fan H, Yao H, Zong J, Chen D, Wang J, Lu C. Comparative genomic analysis shows that Streptococcus suis meningitis isolate SC070731 contains a unique 105K genomic island. Gene 2013; 535:156-64. [PMID: 24316490 DOI: 10.1016/j.gene.2013.11.044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/16/2013] [Accepted: 11/19/2013] [Indexed: 11/29/2022]
Abstract
Streptococcus suis (SS) is an important swine pathogen worldwide that occasionally causes serious infections in humans. SS infection may result in meningitis in pigs and humans. The pathogenic mechanisms of SS are poorly understood. Here, we provide the complete genome sequence of S. suis serotype 2 (SS2) strain SC070731 isolated from a pig with meningitis. The chromosome is 2,138,568bp in length. There are 1933 predicted protein coding sequences and 96.7% (57/59) of the known virulence-associated genes are present in the genome. Strain SC070731 showed similar virulence with SS2 virulent strains HA9801 and ZY05719, but was more virulent than SS2 virulent strain P1/7 in the zebrafish infection model. Comparative genomic analysis revealed a unique 105K genomic island in strain SC070731 that is absent in seven other sequenced SS2 strains. Further analysis of the 105K genomic island indicated that it contained a complete nisin locus similar to the nisin U locus in S. uberis strain 42, a prophage similar to S. oralis phage PH10 and several antibiotic resistance genes. Several proteins in the 105K genomic island, including nisin and RelBE toxin-antitoxin system, contribute to the bacterial fitness and virulence in other pathogenic bacteria. Further investigation of newly identified gene products, including four putative new virulence-associated surface proteins, will improve our understanding of SS pathogenesis.
Collapse
Affiliation(s)
- Zongfu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Weixue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Min Tang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Jing Shao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Chen Dai
- Experimental Teaching Center of Life Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Huochun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China
| | - Jie Zong
- Novel Bioinformatics Co., Ltd, Shanghai, China
| | - Dai Chen
- Novel Bioinformatics Co., Ltd, Shanghai, China
| | | | - Chengping Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| |
Collapse
|
36
|
Lü A, Hu X, Wang Y, Shen X, Zhu A, Shen L, Ming Q, Feng Z. Comparative analysis of the acute response of zebrafish Danio rerio skin to two different bacterial infections. JOURNAL OF AQUATIC ANIMAL HEALTH 2013; 25:243-251. [PMID: 24341765 DOI: 10.1080/08997659.2013.829132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Skin is an important innate immune organ in fish; however, little is known about the skin's immune response to infectious pathogens. We conducted a comparative analysis of the acute immune response of Zebrafish Danio rerio skin against gram-positive (Staphylococcus chromogenes) and gram-negative (Citrobacter freundii) bacterial infections. Gene expression profiles induced from the two different infections were identified by microarray hybridization, with many genes demonstrating an acute immune response in the skin. Differentially expressed genes were mainly involved in response to stress and stimulus, complement activation, acute-phase response, and defense and immune response. Compared with transcription patterns of skin from the two infections, a similar innate immunity (e.g., transferrin, coagulation factor, complements, and lectins) was observed but with different acute-phase genes (e.g., ceruloplasmin, alpha-1-microglobulin, vitellogenin, and heat shock protein). These results suggest that the skin of fish plays an important role in the innate immune responses to bacterial infection.
Collapse
Affiliation(s)
- Aijun Lü
- a School of Life Sciences , Jiangsu Normal University , Xuzhou , 221116 , China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Zhang R, Zhang LL, Ye X, Tian YY, Sun CF, Lu MX, Bai JJ. Transcriptome profiling and digital gene expression analysis of Nile tilapia (Oreochromis niloticus) infected by Streptococcus agalactiae. Mol Biol Rep 2013; 40:5657-68. [DOI: 10.1007/s11033-013-2667-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 09/14/2013] [Indexed: 01/01/2023]
|
38
|
Jia B, Lu H, Liu Q, Yin J, Jiang N, Chen Q. Genome-wide comparative analysis revealed significant transcriptome changes in mice after Toxoplasma gondii infection. Parasit Vectors 2013; 6:161. [PMID: 23734932 PMCID: PMC3679772 DOI: 10.1186/1756-3305-6-161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 05/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an intracellular parasite that can modulate host responses and presumably host behavior. Host responses as well as pathogenesis vary depending on the parasite strains that are responsible for infection. In immune competent individuals, T. gondii preferentially infects tissues of the central nervous systems (CNS), which might be an additional factor in certain psychiatric disorders. While in immune-compromised individuals and pregnant women, the parasite can cause life-threatening infections. With the availability of the genome-wide investigation platform, the global responses in gene expression of the host after T. gondii infection can be systematically investigated. METHODS Total RNA of brain tissues and peripheral lymphocytes of BALB/C mice infected with RH and ME 49 strain T. gondii as well as that of healthy mice were purified and converted to cRNA with incorporated Cy5-CTP (experimental samples), or Cy3-CTP (control samples). The labeled cRNA probes were hybridized to the Whole Mouse Genome Microarray. The impact of parasite infection on gene expression in both brain tissues and peripheral lymphocytes were analyzed. Differentially expressed genes were revalidated with real-time quantitative reverse transcriptase-polymerase chain reaction (Q-PCR). RESULTS Data indicated that the genes associated with immunity were up-regulated after infection by the two parasite strains, but significant up-regulation was observed in both brain tissues and peripheral lymphocytes of mice infected with ME49 strain compared to that infected by RH strain. The pathways related to pathogenesis of the nervous system were more significantly up-regulated in mice infected with RH strain. CONCLUSIONS Genetically distinct T. gondii strains showed clear differences in modulation of host pathophysiological and immunological responses in both brain tissue and peripheral lymphocytes. It was likely that some of the host responses to T. gondii infection were universal, but the immune response and CNS reaction were in a strain-specific manner.
Collapse
Affiliation(s)
- Boyin Jia
- Key Laboratory of Zoonosis, Institute of Zoonosis/College of Veterinary Medicine, Jilin University, Xi An Da Lu 5333, Changchun 130062, China.
| | | | | | | | | | | |
Collapse
|
39
|
Zhang H, Ma Z, Li Y, Zheng J, Yi L, Fan H, Lu C. Identification of a novel collagen type І-binding protein from Streptococcus suis serotype 2. Vet J 2013; 197:406-14. [PMID: 23465548 DOI: 10.1016/j.tvjl.2013.01.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/21/2013] [Accepted: 01/23/2013] [Indexed: 11/24/2022]
Abstract
Streptococcus suis, a major pathogen of pigs, is an emerging zoonotic agent that causes meningitis and septic shock. cbp40 is a putative virulent gene that has been identified using suppression subtractive hybridization performed on the virulent S. suis serotype 2 strain HA9801 and the avirulent S. suis serotype 2 strain T15. Based on predicted protein features showing a shared conserved domain with the collagen-binding protein Cna of Staphylococcus aureus, Cbp40 is likely to function as a direct mediator of collagen adhesion. Here, the cbp40 gene was cloned and the recombinant protein purified. Western blotting using swine convalescent sera confirmed its role as an immunogenic protein. Collagen binding activity could be detected by western affinity blot and ELISA. Conversely, deletion of the cbp40 gene reduced bacterial adhesion to HEp-2 cells, capacity for biofilm formation, and virulence in a zebrafish infection model. The response of the bEnd.3 cell line to infection with the S. suis serotype 2 strain ZY05719 and the cbp40-knockout strain was evaluated using gene expression arrays. The differentially expressed genes were involved in inflammatory and immune responses, leukocyte adhesion and heterophilic cell adhesion. Collectively, these data suggest that Cbp40 plays an important role as an extracellular matrix adhesion protein that interacts with host cells during infection.
Collapse
Affiliation(s)
- Hui Zhang
- Key Lab Animal Disease Diagnostic and Immunology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Pathogen recognition and activation of the innate immune response in zebrafish. Adv Hematol 2012; 2012:159807. [PMID: 22811714 PMCID: PMC3395205 DOI: 10.1155/2012/159807] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 04/22/2012] [Indexed: 12/28/2022] Open
Abstract
The zebrafish has proven itself as an excellent model to study vertebrate innate immunity. It presents us with possibilities for in vivo imaging of host-pathogen interactions which are unparalleled in mammalian model systems. In addition, its suitability for genetic approaches is providing new insights on the mechanisms underlying the innate immune response. Here, we review the pattern recognition receptors that identify invading microbes, as well as the innate immune effector mechanisms that they activate in zebrafish embryos. We compare the current knowledge about these processes in mammalian models and zebrafish and discuss recent studies using zebrafish infection models that have advanced our general understanding of the innate immune system. Furthermore, we use transcriptome analysis of zebrafish infected with E. tarda, S. typhimurium, and M. marinum to visualize the gene expression profiles resulting from these infections. Our data illustrate that the two acute disease-causing pathogens, E. tarda and S. typhimurium, elicit a highly similar proinflammatory gene induction profile, while the chronic disease-causing pathogen, M. marinum, induces a weaker and delayed innate immune response.
Collapse
|
41
|
Fittipaldi N, Segura M, Grenier D, Gottschalk M. Virulence factors involved in the pathogenesis of the infection caused by the swine pathogen and zoonotic agent Streptococcus suis. Future Microbiol 2012; 7:259-79. [PMID: 22324994 DOI: 10.2217/fmb.11.149] [Citation(s) in RCA: 319] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Streptococcus suis is a major swine pathogen responsible for important economic losses to the swine industry worldwide. It is also an emerging zoonotic agent of meningitis and streptococcal toxic shock-like syndrome. Since the recent recognition of the high prevalence of S. suis human disease in southeast and east Asia, the interest of the scientific community in this pathogen has significantly increased. In the last few years, as a direct consequence of these intensified research efforts, large amounts of data on putative virulence factors have appeared in the literature. Although the presence of some proposed virulence factors does not necessarily define a S. suis strain as being virulent, several cell-associated or secreted factors are clearly important for the pathogenesis of the S. suis infection. In order to cause disease, S. suis must colonize the host, breach epithelial barriers, reach and survive in the bloodstream, invade different organs, and cause exaggerated inflammation. In this review, we discuss the potential contribution of different described S. suis virulence factors at each step of the pathogenesis of the infection. Finally, we briefly discuss other described virulence factors, virulence factor candidates and virulence markers for which a precise role at specific steps of the pathogenesis of the S. suis infection has not yet been clearly established.
Collapse
Affiliation(s)
- Nahuel Fittipaldi
- Groupe de Recherche sur les Maladies Infectieuses du Porc & Centre de Recherche en Infectiologie Porcine, Faculté de médecine vétérinaire, Université de Montréal, 3200 rue Sicotte, CP5000, St-Hyacinthe, Quebec, J2S 7C6, Canada
| | | | | | | |
Collapse
|
42
|
Uropathogenic Escherichia coli induces serum amyloid a in mice following urinary tract and systemic inoculation. PLoS One 2012; 7:e32933. [PMID: 22427910 PMCID: PMC3299708 DOI: 10.1371/journal.pone.0032933] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 02/03/2012] [Indexed: 12/28/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein involved in the homeostasis of inflammatory responses and appears to be a vital host defense component with protective anti-infective properties. SAA expression remains poorly defined in many tissues, including the urinary tract which often faces bacterial challenge. Urinary tract infections (UTIs) are usually caused by strains of uropathogenic Escherichia coli (UPEC) and frequently occur among otherwise healthy individuals, many of whom experience bouts of recurrent and relapsing infections despite the use of antibiotics. To date, whether SAA is present in the infected urothelium and whether or not the induction of SAA can protect the host against UPEC is unclear. Here we show, using mouse models coupled with immunofluorescence microscopy and quantitative RT-PCR, that delivery of UPEC either directly into the urinary tract via catheterization or systemically via intraperitoneal injection triggers the expression of SAA. As measured by ELISA, serum levels of SAA1/2 were also transiently elevated in response to UTI, but circulating SAA3 levels were only up-regulated substantially following intraperitoneal inoculation of UPEC. In in vitro assays, physiological relevant levels of SAA1/2 did not affect the growth or viability of UPEC, but were able to block biofilm formation by the uropathogens. We suggest that SAA functions as a critical host defense against UTIs, preventing the formation of biofilms both upon and within the urothelium and possibly providing clinicians with a sensitive serological marker for UTI.
Collapse
|
43
|
Rong J, Zhang W, Wang X, Fan H, Lu C, Yao H. Identification of candidate susceptibility and resistance genes of mice infected with Streptococcus suis type 2. PLoS One 2012; 7:e32150. [PMID: 22384161 PMCID: PMC3288072 DOI: 10.1371/journal.pone.0032150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 01/23/2012] [Indexed: 11/29/2022] Open
Abstract
Streptococcus suis type 2 (SS2) is an important swine pathogen and zoonosis agent. A/J mice are significantly more susceptible than C57BL/6 (B6) mice to SS2 infection, but the genetic basis is largely unknown. Here, alterations in gene expression in SS2 (strain HA9801)-infected mice were identified using Illumina mouse BeadChips. Microarray analysis revealed 3,692 genes differentially expressed in peritoneal macrophages between A/J and B6 mice due to SS2 infection. Between SS2-infected A/J and control A/J mice, 2646 genes were differentially expressed (1469 upregulated; 1177 downregulated). Between SS2-infected B6 and control B6 mice, 1449 genes were differentially expressed (778 upregulated; 671 downregulated). These genes were analyzed for significant Gene Ontology (GO) categories and signaling pathways using the Kyoto Encylopedia of Genes and Genomes (KEGG) database to generate a signaling network. Upregulated genes in A/J and B6 mice were related to response to bacteria, immune response, positive regulation of B cell receptor signaling pathway, type I interferon biosynthesis, defense and inflammatory responses. Additionally, upregulated genes in SS2-infected B6 mice were involved in antigen processing and presentation of exogenous peptides, peptide antigen stabilization, lymphocyte differentiation regulation, positive regulation of monocyte differentiation, antigen receptor-mediated signaling pathway and positive regulation of phagocytosis. Downregulated genes in SS2-infected B6 mice played roles in glycolysis, carbohydrate metabolic process, amino acid metabolism, behavior and muscle regulation. Microarray results were verified by quantitative real-time PCR (qRT-PCR) of 14 representative deregulated genes. Four genes differentially expressed between SS2-infected A/J and B6 mice, toll-like receptor 2 (Tlr2), tumor necrosis factor (Tnf), matrix metalloproteinase 9 (Mmp9) and pentraxin 3 (Ptx3), were previously implicated in the response to S. suis infection. This study identified candidate genes that may influence susceptibility or resistance to SS2 infection in A/J and B6 mice, providing further validation of these models and contributing to understanding of S. suis pathogenic mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Huochun Yao
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
- * E-mail:
| |
Collapse
|
44
|
Wang Y, Liu C, Fang Y, Liu X, Li W, Liu S, Liu Y, Liu Y, Charreyre C, Audonnet JC, Chen P, He Q. Transcription analysis on response of porcine alveolar macrophages to Haemophilus parasuis. BMC Genomics 2012; 13:68. [PMID: 22330747 PMCID: PMC3296652 DOI: 10.1186/1471-2164-13-68] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 02/13/2012] [Indexed: 12/20/2022] Open
Abstract
Background Haemophilus parasuis (H. parasuis) is the etiological agent of Glässer's disease in pigs. Currently, the molecular basis of this infection is largely unknown. The innate immune response is the first line of defense against the infectious disease. Systematical analysis on host innate immune response to the infection is important for understanding the pathogenesis of the infectious microorganisms. Results A total of 428 differentially expressed (DE) genes were identified in the porcine alveolar macrophages (PAMs) 6 days after H. parasuis infection. These genes were principally related to inflammatory response, immune response, microtubule polymerization, regulation of transcript and signal transduction. Through the pathway analysis, the significant pathways mainly concerned with cell adhesion molecules, cytokine-cytokine receptor interaction, complement and coagulation cascades, toll-like receptor signaling pathway, MAPK signaling pathway, suggesting that the host took different strategies to activate immune and inflammatory response upon H. parasuis infection. The global interactions network and two subnetworks of the proteins encoded by DE genes were analyzed by using STRING. Further immunostimulation analysis indicated that mRNA levels of S100 calcium-binding protein A4 (S100A4) and S100 calcium-binding protein A6 (S100A6) in porcine PK-15 cells increased within 48 h and were sustained after administration of lipopolysaccharide (LPS) and Poly (I:C) respectively. The s100a4 and s100a6 genes were found to be up-regulated significantly in lungs, spleen and lymph nodes in H. parasuis infected pigs. We firstly cloned and sequenced the porcine coronin1a gene. Phylogenetic analysis showed that poCORONIN 1A belonged to the group containing the Bos taurus sequence. Structural analysis indicated that the poCORONIN 1A contained putative domains of Trp-Asp (WD) repeats signature, Trp-Asp (WD) repeats profile and Trp-Asp (WD) repeats circular profile at the N-terminus. Conclusions Our present study is the first one focusing on the response of porcine alveolar macrophages to H. parasuis. Our data demonstrate a series of genes are activated upon H. parasuis infection. The observed gene expression profile could help screening the potential host agents for reducing the prevalence of H. parasuis and further understanding the molecular pathogenesis associated with H. parasuis infection in pigs.
Collapse
Affiliation(s)
- Yang Wang
- State key Laboratory of Agricultural Microbiology, Division of Animal Infectious Disease, Huazhong Agricultural University, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lü A, Hu X, Xue J, Zhu J, Wang Y, Zhou G. Gene expression profiling in the skin of zebrafish infected with Citrobacter freundii. FISH & SHELLFISH IMMUNOLOGY 2012; 32:273-283. [PMID: 22155693 DOI: 10.1016/j.fsi.2011.11.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 11/08/2011] [Accepted: 11/18/2011] [Indexed: 05/31/2023]
Abstract
Skin is considered the largest immunologically active organ, but its molecular mechanism remains unclear in fish. Here, Affymetrix Zebrafish GeneChip was used to assess gene expression in the skin of zebrafish (Danio rerio) infected with the bacterium Citrobacter freundii. The results showed that 229 genes were differentially expressed, of which 196 genes were upregulated and 33 genes were downregulated. Gene Ontology and KEGG pathway analyses indicated 88 genes significantly associated with skin immunity involved in complement activation and acute phase response, defense and immune response, response to stress and stimulus, antigen processing and presentation, cell adhesion and migration, platelet activation and coagulation factors, regulation of autophagy and apoptosis. When compared with transcriptional profiles of previously reported carp (Cyprinus carpio) skin, a similar innate immunity (e.g., interferon, lectin, heat shock proteins, complements), and several different acute phase proteins (transferrin, ceruloplasmin, vitellogenin and alpha-1-microglobulin, etc.) were detected in zebrafish skin. The validity of the microarray results was verified by quantitative real-time PCR analysis of nine representative genes. This is first report that skin play important roles in innate immune responses to bacterial infection, which contribute to understanding the defense mechanisms of the skin in fish.
Collapse
Affiliation(s)
- Aijun Lü
- School of Life Sciences, Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, Xuzhou Normal University, Xuzhou 221116, China.
| | | | | | | | | | | |
Collapse
|
46
|
Novoa B, Figueras A. Zebrafish: model for the study of inflammation and the innate immune response to infectious diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:253-75. [PMID: 21948373 DOI: 10.1007/978-1-4614-0106-3_15] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The zebrafish (Danio rerio) has been extensively used in biomedical research as a model to study vertebrate development and hematopoiesis and recently, it has been adopted into varied fields including immunology. After fertilization, larvae survive with only the innate immune responses because adaptive immune system is morphologically and functionally mature only after 4-6 weeks postfertilization. This temporal separation provides a suitable system to study the vertebrate innate immune response in vivo, independently from the adaptive immune response. The transparency of early life stages allows a useful real-time visualization. Adult zebrafish which have complete (innate and adaptative) immune systems offer also advantages over other vertebrate infection models: small size, relatively rapid life cycle, ease of breeding, and a growing list of molecular tools for the study of infectious diseases. In this review, we have tried to give some examples of the potential of zebrafish as a valuable model in innate immunity and inflammation studies.
Collapse
Affiliation(s)
- Beatriz Novoa
- Instituto de Investigaciones Marinas, CSIC, Eduardo Cabello 6, 36208 Vigo, Spain.
| | | |
Collapse
|
47
|
Characterization and functional analysis of atl, a novel gene encoding autolysin in Streptococcus suis. J Bacteriol 2012; 194:1464-73. [PMID: 22228730 DOI: 10.1128/jb.06231-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus suis serotype 2 (S. suis 2) is an important swine and human pathogen responsible for septicemia and meningitis. A novel gene, designated atl and encoding a major autolysin of S. suis 2 virulent strain HA9801, was identified and characterized in this study. The Atl protein contains 1,025 amino acids with a predicted molecular mass of 113 kDa and has a conserved N-acetylmuramoyl-l-alanine amidase domain. Recombinant Atl was expressed in Escherichia coli, and its bacteriolytic and fibronectin-binding activities were confirmed by zymography and Western affinity blotting. Two bacteriolytic bands were shown in the sodium dodecyl sulfate extracts of HA9801, while both were absent from the atl inactivated mutant. Cell chains of the mutant strain became longer than that of the parental strain. In the autolysis assay, HA9801 decreased to 20% of the initial optical density (OD) value, while the mutant strain had almost no autolytic activity. The biofilm capacity of the atl mutant was reduced ∼30% compared to the parental strain. In the zebrafish infection model, the 50% lethal dose of the mutant strain was increased up to 5-fold. Furthermore, the adherence to HEp-2 cells of the atl mutant was 50% less than that of the parental strain. Based on the functional analysis of the recombinant Atl and observed effects of atl inactivation on HA9801, we conclude that Atl is a major autolysin of HA9801. It takes part in cell autolysis, separation of daughter cells, biofilm formation, fibronectin-binding activity, cell adhesion, and pathogenesis of HA9801.
Collapse
|
48
|
Nakao M, Tsujikura M, Ichiki S, Vo TK, Somamoto T. The complement system in teleost fish: progress of post-homolog-hunting researches. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1296-1308. [PMID: 21414344 DOI: 10.1016/j.dci.2011.03.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/05/2011] [Accepted: 03/06/2011] [Indexed: 05/30/2023]
Abstract
Studies on the complement system of bony fish are now finishing a stage of homologue-hunting identification of the components, unveiling existence of almost all the orthologues of mammalian complement components in teleost. Genomic and transcriptomic data for several teleost species have contributed much for the homologue-hunting research progress. Only an exception is identification of orthologues of mammalian complement regulatory proteins and complement receptors. It is of particular interest that teleost complement components often exist as multiple isoforms with possible functional divergence. This review summarizes research progress of teleost complement system following the molecular identification and sequence analysis of the components. The findings of extensive expression analyses of the complement components with special emphasis of their prominent extrahepatic expression, acute-phase response to immunostimulation and various microbial infections, and ontogenic development including maternal transfer are discussed to infer teleost-specific functions of the complement system. Importance of the protein level characterization of the complement components is also emphasized, especially for understanding of the isotypic diversity of the components, a unique feature of teleost complement system.
Collapse
Affiliation(s)
- Miki Nakao
- Laboratory of Marine Biochemistry, Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan.
| | | | | | | | | |
Collapse
|
49
|
Boltaña S, Roher N, Goetz FW, Mackenzie SA. PAMPs, PRRs and the genomics of gram negative bacterial recognition in fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1195-1203. [PMID: 21453721 DOI: 10.1016/j.dci.2011.02.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 01/12/2011] [Accepted: 02/25/2011] [Indexed: 05/30/2023]
Abstract
Understanding the mechanisms that underpin pathogen recognition and subsequent orchestration of the immune response in fish is an area of significant importance for both basic research and management of health in aquaculture. In recent years much attention has been given to the identification of pattern recognition receptors (PRRs) in fish, however, characterisation of interactions with specific pathogen-associated molecular patterns (PAMPs) is still incomplete. Microarray studies have significantly contributed to functional studies and early descriptions of PAMP-PRR driven activation of specific response cassettes in the genome have been obtained although much is left to be done. In this review we will address gram negative (G-negative) bacterial recognition in fish addressing contributing factors such as structure-function relationships between G-negative PAMPs, current knowledge of fish PRRs and the input achieved by microarray-based studies ranging from in vivo infection studies to directed in vitro PAMP-cell studies. Finally we revisit the endotoxic recognition paradigm in fish and suggest a series of future perspectives that could contribute toward the further elucidation of G-negative bacterial recognition across the highly diverse group of vertebrates that encompass the fishes.
Collapse
Affiliation(s)
- Sebastian Boltaña
- Institute of Biotechnology and Biomedicine, Dep. Biologia Cel·lular, Immunologia i Fisiologia Animal, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | | | | | | |
Collapse
|
50
|
Borst LB, Patterson SK, Lanka S, Suyemoto MM, Maddox CW. Zebrafish (Danio rerio) as a screen for attenuation of Lancefield group C streptococci and a model for streptococcal pathogenesis. Vet Pathol 2011; 50:457-67. [PMID: 21997564 DOI: 10.1177/0300985811424731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Group C streptococci are highly contagious pyogenic bacteria responsible for respiratory tract, lymph node, urogenital tract, and wound infections. Wild-type strains of Streptococcus equi ssp equi (S. equi) and Streptococcus equi ssp zooepidemicus (S. zoo) as well as a commercially available modified live vaccine strain of S. equi were evaluated for virulence in zebrafish. Survival times, histologic lesions, and relative gene expression were compared among groups. Based on the intramuscular route of infection, significantly shorter survival times were observed in fish infected with wild-type strain when compared to modified live vaccine and S. zoo strains. Histologically, S. zoo-infected fish demonstrated a marked increase in inflammatory infiltrates (predominantly macrophages) at the site of infection, as well as increased cellularity in the spleen and renal interstitium. In contrast, minimal cellular immune response was observed in S. equi-injected fish with local tissue necrosis and edema predominating. Based on whole comparative genomic hybridization, increased transcription of positive acute-phase proteins, coagulation factors, and antimicrobial peptides were observed in S. equi-injected fish relative to S. zoo-injected fish, while mediators of cellular inflammation, including CXC chemokines and granulin, were upregulated in S. zoo-injected fish relative to S. equi-injected fish. In a screen of 11 clinical isolates, S. equi strains with a single nucleotide deletion in the upstream region of szp, a known virulence factor of streptococci, were found to be significantly attenuated in zebrafish. These collective findings underscore the value of the zebrafish as a model of streptococcal pathogenesis.
Collapse
Affiliation(s)
- L B Borst
- North Carolina State University, College of Veterinary Medicine, 4700 Hillsborough Street, Raleigh, NC 27606, USA.
| | | | | | | | | |
Collapse
|