1
|
Sun D, Gou H, Zhang Y, Li J, Dai C, Shen H, Chen K, Wang Y, Pan P, Zhu T, Xu C, Shan T, Liao M, Zhang J. Salmonella Typhimurium persistently infects host via its effector SseJ-induced PHB2-mediated mitophagy. Autophagy 2025; 21:1228-1244. [PMID: 39902787 DOI: 10.1080/15548627.2025.2462511] [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: 04/27/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Despite decades of research on effective methods to resist Salmonella enterica serovar Typhimurium (S. Typhimurium) pathogenicity, the mechanisms of S. Typhimurium-host interactions have not been fully determined. S. Typhimurium is characterized as an important zoonosis in public health worldwide because of its endemicity, high morbidity, and difficulty in applying control and prevention measures. Herein, we introduce a novel bacterial factor, secretion system effector J (SseJ), and its interactive host protein, PHB2 (prohibitin 2). We explored whether SseJ affected S. Typhimurium replication and survival in the host. S. Typhimurium infection caused severe mitochondrial damage and mitophagy, which facilitated S. Typhimurium proliferation in cells. S. Typhimurium SseJ activated the PINK1 (PTEN induced kinase 1)-PRKN (parkin RBR E3 ubiquitin protein ligase)-autophagosome-dependent mitophagy pathway, aided by the mitophagy receptor PHB2, for bacterial survival and persistent infection. Moreover, suppression of mitophagy alleviated the pathogenicity of S. Typhimurium. In conclusion, S. Typhimurium infection could be antagonized by targeting the SseJ-PHB2-mediated host mitochondrial autophagy pathway.Abbreviation: ACTB: actin beta; BafA1: bafilomycin A1; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; co-IP: co-immunoprecipitation; CFU: colony-forming units; COX4/COXIV: cytochrome c oxidase subunit 4; CQ: chloroquine; hpi: h post-bacterial infection; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; Mdivi-1:mitophagy inhibitor mitochondrial division inhibitor 1; MFN2: mitofusin 2; MG132: z-leu-leu-leucinal; MOI: multiplicity of infection; mtDNA: mitochondrial DNA; PBS: phosphate-buffered saline; PGAM5: PGAM family member 5, mitochondrial serine/threonine protein phosphatase; PHB2: prohibitin 2; PINK1: PTEN induced kinase 1; qPCR: quantitative real-time reverse transcription PCR; Roc-A: Rocaglamide A; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; SCVs: Salmonella-containing vacuoles; siRNA: small interfering RNA; SPI-2: Salmonella pathogenicity island 2; SseJ: secretion system effector J; S. Typhimurium: Salmonella enterica serovar Typhimurium; S.T-ΔSseJ: SseJ gene-deleted Salmonella Typhimurium strains; S.T-CΔSseJ: SseJ-complemented Salmonella Typhimurium strains; WT: wild-type.
Collapse
Affiliation(s)
- Dage Sun
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongchao Gou
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Yu Zhang
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai, China
| | - Jiayi Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Changzhi Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Haiyan Shen
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Kaifeng Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yu Wang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Peng Pan
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ting Zhu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chenggang Xu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Tongling Shan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Zhongkai University of Agricultural Engineering, Guangzhou, China
| | - Jianmin Zhang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
2
|
Almufarriji FM, Alotaibi BS, Alamri AS, S Alkhalil S, Alwethaynani MS. Phytoconstituents from Artemisia annua medicinal plant as potent inhibitors targeting Salmonella SpvB: a molecular docking and dynamic study. J Biomol Struct Dyn 2025:1-14. [PMID: 40256876 DOI: 10.1080/07391102.2025.2492237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Salmonella, a genus with a global presence, is a leading cause of diarrheal diseases in both humans and animals. With over 2,400 distinct serotypes, most exhibiting minimal host specificity, Salmonella infection remains a significant public health issue. It poses a substantial economic burden on both developed and developing nations due to the costs associated with disease surveillance, prevention, and treatment. To address this global challenge, it is essential to explore cost-effective therapeutic interventions derived from medicinal plants. In this study, we targeted the Salmonella SpvB ATR domain for molecular docking of phytochemical compounds. A library of 392 phytochemical compounds from the Artemisia annua (Sweet wormwood) medicinal plant was utilized. In the initial screening, the top 20 phytochemical compounds were selected based on their high binding affinity toward SpvB. These 20 compounds underwent interaction analysis, revealing that two compounds, IMPHY004808 and IMPHY015047, formed crucial interactions. The IMPHY004808 compound bound at binding site residues ARG414, ARG471, LEU473, and GLU538, with residue SER501 present at the active site. Similarly, the IMPHY015047 compound formed bonds at binding site residues ARG471, ARG414, GLY472, and GLU538, while residue SER501 was present at the active site of SpvB. The trajectory analysis of 500 ns MD simulation, including deviation, fluctuation, compactness, surface area calculation, secondary structure element alterations, and hydrogen bond analysis, showed that the complexes were stable during the simulation time. Moreover, PCA with minimal vibration, FEL analysis and MMPBSA analysis strongly recommend that the complexes were stable and further validation with experimentation is needed.
Collapse
Affiliation(s)
- Fawaz M Almufarriji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Ahlam Saleh Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Samia S Alkhalil
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Maher S Alwethaynani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Nicastro GG, Sibinelli-Sousa S, Hespanhol JT, Santos TWC, Munoz JP, Santos RS, Perez-Sepulveda BM, Miyamoto S, Aravind L, de Souza RF, Bayer-Santos E. Identification of novel toxin domains and characterization of a broadly distributed family of lipid-targeting NlpC/P60. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615498. [PMID: 40236209 PMCID: PMC11996579 DOI: 10.1101/2024.09.27.615498] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Bacterial warfare is a common and ancient phenomenon in nature, where bacterial species use strategies to inhibit the growth or kill competitors. This involves the production and deployment of antibacterial toxins that disrupt essential cellular processes in target cells. Polymorphic toxins comprise a group of offensive systems with a modular structure featuring a conserved N-terminal translocation domain fused to diverse C-terminal toxin domains. The continuous arms race in which bacteria acquire new toxin and immunity proteins to promote increased adaptation to their environment is responsible for the diversification of this toxin repertoire. Here, we deployed in-silico strategies to analyze 10,000 genomes and identify toxin domains secreted via the type VI secretion system of Salmonella . We identified and manually curated 128 candidates, which are widespread polymorphic toxins detected in a vast array of species and linked to diverse secretion systems. In addition, 45 previously uncharacterized toxin domains were identified. STox15 was among the most frequent candidates found in the dataset and was selected for in-depth characterization. STox15 is an antibacterial effector belonging to the NlpC/P60 papain-like fold superfamily with a permuted catalytic core typical of lipid-targeting versions rather than peptidases or amidases. Biochemical analysis with recombinant protein and lipidomics of intoxicated Escherichia coli revealed that STox15 displays phospholipase activity cleaving off acyl groups from phosphatidylglycerol and phosphatidylethanolamine. Importance This work broadens our understanding of polymorphic toxin domains and provides the first direct characterization of a lipid-targeting NlpC/P60 domain in biological conflicts.
Collapse
|
4
|
Bansal G, Ghanem M, Sears KT, Galen JE, Tennant SM. Genetic engineering of Salmonella spp. for novel vaccine strategies and therapeutics. EcoSal Plus 2024; 12:eesp00042023. [PMID: 39023252 PMCID: PMC11636237 DOI: 10.1128/ecosalplus.esp-0004-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
Salmonella enterica is a diverse species that infects both humans and animals. S. enterica subspecies enterica consists of more than 1,500 serovars. Unlike typhoidal Salmonella serovars which are human host-restricted, non-typhoidal Salmonella (NTS) serovars are associated with foodborne illnesses worldwide and are transmitted via the food chain. Additionally, NTS serovars can cause disease in livestock animals causing significant economic losses. Salmonella is a well-studied model organism that is easy to manipulate and evaluate in animal models of infection. Advances in genetic engineering approaches in recent years have led to the development of Salmonella vaccines for both humans and animals. In this review, we focus on current progress of recombinant live-attenuated Salmonella vaccines, their use as a source of antigens for parenteral vaccines, their use as live-vector vaccines to deliver foreign antigens, and their use as therapeutic cancer vaccines in humans. We also describe development of live-attenuated Salmonella vaccines and live-vector vaccines for use in animals.
Collapse
Affiliation(s)
- Garima Bansal
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mostafa Ghanem
- Department of Veterinary Medicine, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Khandra T. Sears
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James E. Galen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Bao X, Wu J. Natural anti-adhesive components against pathogenic bacterial adhesion and infection in gastrointestinal tract: case studies of Helicobacter pylori, Salmonella enterica, Clostridium difficile, and diarrheagenic Escherichia coli. Crit Rev Food Sci Nutr 2024:1-46. [PMID: 39666022 DOI: 10.1080/10408398.2024.2436139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Antimicrobial resistance (AMR) poses a global public health concern. Recognizing the critical role of bacterial adhesion in pathogenesis of infection, anti-adhesive therapy emerges as a promising approach to impede initial bacterial attachment, thus preventing pathogenic colonization and infection. Natural anti-adhesive agents derived from food sources are generally safe and have the potential to inhibit the emergence of resistant bacteria. This comprehensive review explored diverse natural dietary components exhibiting anti-adhesive activities against several model enteric pathogens, including Helicobacter pylori, Salmonella enterica, Clostridium difficile, and three key diarrheagenic Escherichia coli (i.e., enterotoxigenic E. coli, enteropathogenic E. coli, and enterohemorrhagic E. coli). Investigating various anti-adhesive products will advance our understanding of current research of the field and inspire further development of these agents as potential nutraceuticals or adjuvants to improve the efficacy of conventional antibiotics.
Collapse
Affiliation(s)
- Xiaoyu Bao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Kim S, Lee YH. The Salmonella enterica EnvE is an Outer Membrane Lipoprotein and Its Gene Expression Leads to Transcriptional Repression of the Virulence Gene msgA. J Microbiol 2024; 62:1013-1022. [PMID: 39546166 DOI: 10.1007/s12275-024-00183-4] [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: 02/14/2024] [Revised: 09/18/2024] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
The envE gene of Salmonella enterica serovar Typhimurium is encoded within Salmonella Pathogenicity Island-11 (SPI-11) and is located immediately upstream of the virulence gene msgA (macrophage survival gene A) in the same transcriptional orientation. To date, the characteristics and roles of envE remain largely unexplored. In this study, we show that EnvE, a predicted lipoprotein, is localized on the outer membrane using sucrose gradient ultracentrifugation. Under oxidative stress conditions, envE transcription is suppressed, while msgA transcription is induced, indicating an inverse correlation between the mRNA levels of the two neighboring genes. Importantly, inactivation of envE leads to constitutive transcription of msgA regardless of the presence of oxidative stress. Moreover, trans-complementation of the envE mutant with a plasmid-borne envE fails to prevent the induction of msgA transcription, suggesting that envE functions as a cis-regulatory element rather than a trans-acting factor. We further show that both inactivation and complementation of envE confer wild-type levels of resistance to oxidative stress by ensuring the expression of msgA. Our data suggest that the S. enterica envE gene encodes an outer membrane lipoprotein, and its transcription represses msgA expression in a cis-acting manner, probably by transcriptional interference, although the exact molecular details are yet unclear.
Collapse
Affiliation(s)
- Sinyeon Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
- MThera Pharma Co., Seoul, 07793, Republic of Korea
| | - Yong Heon Lee
- Department of Biomedical Laboratory Science, Dongseo University, Busan, 47011, Republic of Korea.
| |
Collapse
|
7
|
Zeng Y, Shen M, Liu S, Zhou X. Characterization and resistance mechanism of phage-resistant strains of Salmonella enteritidis. Poult Sci 2024; 103:103756. [PMID: 38652948 PMCID: PMC11063523 DOI: 10.1016/j.psj.2024.103756] [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: 02/07/2024] [Revised: 03/25/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024] Open
Abstract
In the face of the increasingly severe problem of antibiotic resistance, phage therapy is regarded as a highly potential alternative. Compared with traditional antimicrobial agents, a key research area of phage therapy is the study of phage-resistant mutant bacteria. To effectively monitor and prevent this resistance, it is crucial to conduct in-depth exploration of the mechanism behind phage resistance. In this study, a strain of Salmonella enteritidis (sm140) and the corresponding phage (Psm140) were isolated from chicken liver and sewage, respectively. Using the double-layer plate method, successfully screened out phage-resistant mutant strains. Whole-genome resequencing of 3 resistant strains found that the wbaP gene of all 3 strains had mutations at a specific position (1,118), with the base changing from G to A. This mutation causes the gene-encoded glycine to be replaced by aspartic acid. Subsequent studies found that the frequency of this gene mutation is extremely high, reaching 84%, and all mutations occur at the same position. To further explore the relationship between the wbaP gene and phage resistance, knockout strains and complement strains of the wbaP gene were constructed. The experimental results confirmed the association between the wbaP gene and phage resistance. At the same time, biological characteristics and virulence were evaluated for wild strains, resistant strains, knockout strains, and complement strains. It was found that mutations or deletions of the wbaP gene lead to a decrease in bacterial environmental adaptability and virulence. Through systematic research on the mechanism and biological characteristics of phage resistance, this study provides important references and guidance for the development of new phage therapies, promoting progress in the field of antimicrobial treatment. At the same time, the emergence of phage resistance due to wbaP gene mutations is reported for the first time in salmonella, providing a new perspective and ideas for further studying phage resistance mechanisms.
Collapse
Affiliation(s)
- Yukun Zeng
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Mangmang Shen
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Shenglong Liu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xin Zhou
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
8
|
Medvedev KE, Zhang J, Schaeffer RD, Kinch LN, Cong Q, Grishin NV. Structure classification of the proteins from Salmonella enterica pangenome revealed novel potential pathogenicity islands. Sci Rep 2024; 14:12260. [PMID: 38806511 PMCID: PMC11133325 DOI: 10.1038/s41598-024-60991-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
Salmonella enterica is a pathogenic bacterium known for causing severe typhoid fever in humans, making it important to study due to its potential health risks and significant impact on public health. This study provides evolutionary classification of proteins from Salmonella enterica pangenome. We classified 17,238 domains from 13,147 proteins from 79,758 Salmonella enterica strains and studied in detail domains of 272 proteins from 14 characterized Salmonella pathogenicity islands (SPIs). Among SPIs-related proteins, 90 proteins function in the secretion machinery. 41% domains of SPI proteins have no previous sequence annotation. By comparing clinical and environmental isolates, we identified 3682 proteins that are overrepresented in clinical group that we consider as potentially pathogenic. Among domains of potentially pathogenic proteins only 50% domains were annotated by sequence methods previously. Moreover, 36% (1330 out of 3682) of potentially pathogenic proteins cannot be classified into Evolutionary Classification of Protein Domains database (ECOD). Among classified domains of potentially pathogenic proteins the most populated homology groups include helix-turn-helix (HTH), Immunoglobulin-related, and P-loop domains-related. Functional analysis revealed overrepresentation of these protein in biological processes related to viral entry into host cell, antibiotic biosynthesis, DNA metabolism and conformation change, and underrepresentation in translational processes. Analysis of the potentially pathogenic proteins indicates that they form 119 clusters or novel potential pathogenicity islands (NPPIs) within the Salmonella genome, suggesting their potential contribution to the bacterium's virulence. One of the NPPIs revealed significant overrepresentation of potentially pathogenic proteins. Overall, our analysis revealed that identified potentially pathogenic proteins are poorly studied.
Collapse
Affiliation(s)
- Kirill E Medvedev
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Jing Zhang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - R Dustin Schaeffer
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa N Kinch
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qian Cong
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
9
|
Wang X, Zhu H, Hu J, Zhang B, Guo W, Wang Z, Wang D, Qi J, Tian M, Bao Y, Si F, Wang S. Genetic distribution, characterization, and function of Escherichia coli type III secretion system 2 (ETT2). iScience 2024; 27:109763. [PMID: 38706860 PMCID: PMC11068852 DOI: 10.1016/j.isci.2024.109763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024] Open
Abstract
Many Gram-negative bacteria use type Ⅲ secretion system (T3SS) to inject effector proteins and subvert host signaling pathways, facilitating the growth, survival, and virulence. Notably, some bacteria harbor multiple distinct T3SSs with different functions. An extraordinary T3SS, the Escherichia coli Type III Secretion System 2 (ETT2), is widespread among Escherichia coli (E. coli) strains. Since many ETT2 carry genetic mutations or deletions, it is thought to be nonfunctional. However, increasing studies highlight ETT2 contributes to E. coli pathogenesis. Here, we present a comprehensive overview of genetic distribution and characterization of ETT2. Subsequently, we outline its functional potential, contending that an intact ETT2 may retain the capacity to translocate effector proteins and manipulate the host's innate immune response. Given the potential zoonotic implications associated with ETT2-carrying bacteria, further investigations into the structure, function and regulation of ETT2 are imperative for comprehensive understanding of E. coli pathogenicity and the development of effective control strategies.
Collapse
Affiliation(s)
- Xinyu Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Hong Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Jiangang Hu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Beibei Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Weiqi Guo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Zhiyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Di Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Jingjing Qi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Mingxing Tian
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Yanqing Bao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| |
Collapse
|
10
|
Brek T, Gohal GA, Yasir M, Azhar EI, Al-Zahrani IA. Meningitis and Bacteremia by Unusual Serotype of Salmonella enterica Strain: A Whole Genome Analysis. Interdiscip Perspect Infect Dis 2024; 2024:3554734. [PMID: 38558876 PMCID: PMC10980553 DOI: 10.1155/2024/3554734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/27/2023] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Background Although meningitis caused by Salmonella species is relatively rare and accounts for <1% of the confirmed cases in neonates, it is associated with case complications and fatality rates up to 50-70% when compared to other forms of Gram-negative bacilli meningitis. Objectives We conducted an investigation into the first reported case of neonatal meningitis caused by nontyphoidal S. enterica in Jazan, a region in the southwestern part of Saudi Arabia. Methods CSF and blood culture were collected from a female neonate patient to confirm the presence of bacterial meningitis. WGS was conducted to find out the comprehensive genomic characterization of S. enterica isolate. Results A 3-week-old infant was admitted to a local hospital with fever, poor feeding, and hypoactivity. She was diagnosed with Salmonella meningitis and bacteremia caused by S. enterica, which was sensitive to all antimicrobials tested. WGS revealed the specific strain to be S. enterica serotype Johannesburg JZ01, belonging to ST515 and cgMLST 304742. Conclusions We presented a genomic report of rare case of NTS meningitis in an infant who is living in a rural town in Jazan region, Saudi Arabia. Further research is required to understand the impact of host genetic factors on invasive nontyphoidal Salmonella infection.
Collapse
Affiliation(s)
- Thamer Brek
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Public Health Laboratory, The Regional Laboratory and the Central Blood Bank, Jazan Health Directorate, Jazan, Saudi Arabia
| | - Gassem A. Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Muhammad Yasir
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam I. Azhar
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibrahim A. Al-Zahrani
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Mumtaz MN, Irfan M, Siraj S, Khan A, Khan H, Imran M, Khan IA, Khan A. Whole-genome sequencing of extensively drug-resistant Salmonella enterica serovar Typhi clinical isolates from the Peshawar region of Pakistan. J Infect Public Health 2024; 17:271-282. [PMID: 38134602 DOI: 10.1016/j.jiph.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/18/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Typhoid fever, caused by Salmonella enterica serovar Typhi, is a significant public health concern due to the escalating of antimicrobial resistance (AMR), with limited treatment options for extensively drug-resistant (XDR) S. Typhi strains pose a serious threat to disease management and control. This study aimed to investigate the genomic characteristics, epidemiology and AMR genes of XDR S. Typhi strains from typhoid fever patients in Pakistan. METHODOLOGY We assessed 200 patients with enteric fever symptoms, confirming 65 S. Typhi cases through culturing and biochemical tests. Subsequent antimicrobial susceptibility testing revealed 40 cases of extensively drug-resistant (XDR) and 25 cases of multi-drug resistance (MDR). Thirteen XDR strains were selected for whole-genome sequencing, to analyze their sequence type, phylogenetics, resistance genes, pathogenicity islands, and plasmid sequences using variety of data analysis resources. Pangenome analysis was conducted for 140 XDR strains, including thirteen in-house and 127 strains reported from other regions of Pakistan, to assess their genetic diversity and functional annotation. RESULTS MLST analysis classified all isolates as sequence type 1 (ST-1) with 4.3.1.1. P1 genotype characterization. Prophage and Salmonella Pathogenicity Island (SPI) analysis identified intact prophages and eight SPIs involved in Salmonella's invasion and replication within host cells. Genome data analysis revealed numerous AMR genes including dfrA7, sul1, qnrS1, TEM-1, Cat1, and CTX-M-15, and SNPs associated with antibiotics resistance. IncY, IncQ1, pMAC, and pAbTS2 plasmids, conferring antimicrobial resistance, were detected in a few XDR S. Typhi strains. Phylogenetic analysis inferred a close epidemiological linkage among XDR strains from different regions of Pakistan. Pangenome was noted closed among these strains and functional annotation highlighted genes related to metabolism and pathogenesis. CONCLUSION This study revealed a uniform genotypic background among XDR S. Typhi strains in Pakistan, signifying a persistence transmission of a single, highly antibiotic-resistant clone. The closed pan-genome observed underscores limited genetic diversity and highlights the importance of genomic surveillance for combating drug-resistant typhoid infections.
Collapse
Affiliation(s)
- Mah Noor Mumtaz
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan
| | - Muhammad Irfan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | - Sami Siraj
- Institute of Pharmaceutical Sciences, Khyber Medical University (KMU), Peshawar, Pakistan
| | - Aslam Khan
- Department of Pathology, Medical Teaching Institution MTI, Hayatabad Medical Complex (HMC), Peshawar, Pakistan
| | - Hizbullah Khan
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Muhammad Imran
- Research center for Advanced Materials Science (RCAMS), Chemistry Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Ishtiaq Ahmad Khan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi, Karachi, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan.
| |
Collapse
|
12
|
Pico-Rodríguez JT, Martínez-Jarquín H, Gómez-Chávez JDJ, Juárez-Ramírez M, Martínez-Chavarría LC. Effect of Salmonella pathogenicity island 1 and 2 (SPI-1 and SPI-2) deletion on intestinal colonization and systemic dissemination in chickens. Vet Res Commun 2024; 48:49-60. [PMID: 37490241 PMCID: PMC10811122 DOI: 10.1007/s11259-023-10185-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
Salmonella's virulence genes are located in two regions known as Salmonella pathogenicity islands 1 and 2 (SPI-1 and SPI-2). SPI-1 allows the bacteria to invade the intestine, while SPI-2 is important for intracellular survival and replication, although it is also necessary for intestinal disease. The aim of this study was to evaluate the effect of the deletion of SPI-1 or SPI-2 genes on the intestinal and systemic salmonellosis using the avian model. Groups of chickens were orally infected with 1010 Colony-Forming Units (CFU) of S. Typhimurium SL1344 WT strain, as well as mutants ∆SPI-1 or ∆SPI-2. At different times post-infection, 5 chickens from each group were euthanized and examined postmortem. Cecum and liver were taken from each chicken for determination of CFU's, histopathological analysis and immunochemistry. Bacterial colonies were recovered from the liver and cecum samples infected with WT strain, while in the cultures from the organs infected with the mutant strains no colonies were recovered or were drastically affected in the ability to survive. In histopathological analysis, the WT strain produced lesions in liver and ceca, and it was detected by immunohistochemistry throughout the course of the infection. On the other hand, organs of chickens infected with ∆SPI-1 or ∆SPI-2 showed attenuated lesions and the immunohistochemistry revealed less bacteria compared to the WT strain. Taken together, our results show the importance of SPI-1 and SPI-2 genes for the complete intestinal and systemic disease in an in vivo avian model.
Collapse
Affiliation(s)
- Jwerlly Tatiana Pico-Rodríguez
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México
| | - Hugo Martínez-Jarquín
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México
| | - José de Jesús Gómez-Chávez
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México
| | - Mireya Juárez-Ramírez
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México
| | - Luary Carolina Martínez-Chavarría
- Departamento de Patología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México.
| |
Collapse
|
13
|
Cortés-Avalos D, Borges Farias A, Romero-González LE, Lara-Ochoa C, Villa-Tanaca L, García-Del Portillo F, López-Guerrero V, Bustamante VH, Pérez-Rueda E, Ibarra JA. Interactions between the AraC/XylS-like transcriptional activator InvF of Salmonella Typhimurium, the RNA polymerase alpha subunit and the chaperone SicA. Sci Rep 2024; 14:156. [PMID: 38167847 PMCID: PMC10761746 DOI: 10.1038/s41598-023-50636-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Salmonella enterica serovar Typhimurium causes gastroenteritis and systemic infections in humans. For this bacterium the expression of a type III secretion system (T3SS) and effector proteins encoded in the Salmonella pathogenicity island-1 (SPI-1), is keystone for the virulence of this bacterium. Expression of these is controlled by a regulatory cascade starting with the transcriptional regulators HilD, HilC and RtsA that induce the expression of HilA, which then activates expression of the regulator InvF, a transcriptional regulator of the AraC/XylS family. InvF needs to interact with the chaperone SicA to activate transcription of SPI-1 genes including sicA, sopB, sptP, sopE, sopE2, and STM1239. InvF very likely acts as a classical activator; however, whether InvF interacts with the RNA polymerase alpha subunit RpoA has not been determined. Results from this study confirm the interaction between InvF with SicA and reveal that both proteins interact with the RNAP alpha subunit. Thus, our study further supports that the InvF/SicA complex acts as a classical activator. Additionally, we showed for the first time an interaction between a chaperone of T3SS effectors (SicA) and the RNAP.
Collapse
Affiliation(s)
- Daniel Cortés-Avalos
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prol. Carpio y Plan de Ayala S/N, Col. Santo Tomás 11340, Mexico City, Mexico
| | - André Borges Farias
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica del Estado de Yucatán, Mérida, Mexico
| | - Luis E Romero-González
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prol. Carpio y Plan de Ayala S/N, Col. Santo Tomás 11340, Mexico City, Mexico
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Cristina Lara-Ochoa
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Lourdes Villa-Tanaca
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prol. Carpio y Plan de Ayala S/N, Col. Santo Tomás 11340, Mexico City, Mexico
| | - Francisco García-Del Portillo
- Laboratory of Intracellular Bacterial Pathogens, National Centre for Biotechnology (CNB)-CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Vanessa López-Guerrero
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Víctor H Bustamante
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Ernesto Pérez-Rueda
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica del Estado de Yucatán, Mérida, Mexico
| | - J Antonio Ibarra
- Laboratorio de Genética Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prol. Carpio y Plan de Ayala S/N, Col. Santo Tomás 11340, Mexico City, Mexico.
| |
Collapse
|
14
|
Bandyopadhyay S, Zhang X, Ascura A, Edelblum KL, Bonder EM, Gao N. Salmonella engages CDC42 effector protein 1 for intracellular invasion. J Cell Physiol 2024; 239:36-50. [PMID: 37877586 PMCID: PMC11730249 DOI: 10.1002/jcp.31142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/25/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023]
Abstract
Human enterocytes are primary targets of infection by invasive bacterium Salmonella Typhimurium, and studies using nonintestinal epithelial cells established that S. Typhimurium activates Rho family GTPases, primarily CDC42, to modulate the actin cytoskeletal network for invasion. The host intracellular protein network that engages CDC42 and influences the pathogen's invasive capacity are relatively unclear. Here, proteomic analyses of canonical and variant CDC42 interactomes identified a poorly characterized CDC42 interacting protein, CDC42EP1, whose intracellular localization is rapidly redistributed and aggregated around the invading bacteria. CDC42EP1 associates with SEPTIN-7 and Villin, and its relocalization and bacterial engagement depend on host CDC42 and S. Typhimurium's capability of activating CDC42. Unlike CDC42, CDC42EP1 is not required for S. Typhimurium's initial cellular entry but is found to associate with Salmonella-containing vacuoles after long-term infections, indicating a contribution to the pathogen's intracellular growth and replication. These results uncover a new host regulator of enteric Salmonella infections, which may be targeted to restrict bacterial load at the primary site of infection to prevent systemic spread.
Collapse
Affiliation(s)
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Andrea Ascura
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Karen L. Edelblum
- Department of Pathology, Immunology, and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Edward M. Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
15
|
Joiner JD, Steinchen W, Mozer N, Kronenberger T, Bange G, Poso A, Wagner S, Hartmann MD. HilE represses the activity of the Salmonella virulence regulator HilD via a mechanism distinct from that of intestinal long-chain fatty acids. J Biol Chem 2023; 299:105387. [PMID: 37890783 PMCID: PMC10696396 DOI: 10.1016/j.jbc.2023.105387] [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: 03/19/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The expression of virulence factors essential for the invasion of host cells by Salmonella enterica is tightly controlled by a network of transcription regulators. The AraC/XylS transcription factor HilD is the main integration point of environmental signals into this regulatory network, with many factors affecting HilD activity. Long-chain fatty acids, which are highly abundant throughout the host intestine, directly bind to and repress HilD, acting as environmental cues to coordinate virulence gene expression. The regulatory protein HilE also negatively regulates HilD activity, through a protein-protein interaction. Both of these regulators inhibit HilD dimerization, preventing HilD from binding to target DNA. We investigated the structural basis of these mechanisms of HilD repression. Long-chain fatty acids bind to a conserved pocket in HilD, in a comparable manner to that reported for other AraC/XylS regulators, whereas HilE forms a stable heterodimer with HilD by binding to the HilD dimerization interface. Our results highlight two distinct, mutually exclusive mechanisms by which HilD activity is repressed, which could be exploited for the development of new antivirulence leads.
Collapse
Affiliation(s)
- Joe D Joiner
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Wieland Steinchen
- Center for Synthetic Microbiology, Philipps University of Marburg, Marburg, Germany; Department of Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Nick Mozer
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Thales Kronenberger
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany; Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland; Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany
| | - Gert Bange
- Center for Synthetic Microbiology, Philipps University of Marburg, Marburg, Germany; Department of Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Antti Poso
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany; Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, Tübingen, Germany; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Samuel Wagner
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany; Partner-site Tübingen, German Center for Infection Research (DZIF), Tübingen, Germany
| | - Marcus D Hartmann
- Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany; Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
16
|
Bharathan G, Mundra S, Darwich DM, Saeed MM, Al Hafri ASA, Alsalmi MMSM, Maqsood S, Mudgil P, Fanning S, Srikumar S. Regulation of iron metabolism is critical for the survival of Salmonella Typhimurium in pasteurized milk. Food Microbiol 2023; 115:104326. [PMID: 37567619 DOI: 10.1016/j.fm.2023.104326] [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: 02/06/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 08/13/2023]
Abstract
Salmonella is known to survive in raw/pasteurized milk and cause foodborne outbreaks. Lactoferrin, present in milk from all animal sources, is an iron-binding glycoprotein that limits the availability of iron to pathogenic bacteria. Despite the presence of lactoferrins, Salmonella can grow in milk obtained from different animal sources. However, the mechanism by which Salmonella overcomes iron scarcity induced by lactoferrin in milk is not evaluated yet. Salmonella employs the DNA binding transcriptional regulator Fur (ferric update regulator) to mediate iron uptake during survival in iron deplete conditions. To understand the importance of Fur in Salmonella milk growth, we profiled the growth of Salmonella Typhimurium Δfur (ST4/74Δfur) in both bovine and camel milk. ST4/74Δfur was highly inhibited in milk compared to wild-type ST4/74, confirming the importance of Fur mediated regulation of iron metabolism in Salmonella milk growth. We further studied the biology of ST4/74Δfur to understand the importance of iron metabolism in Salmonella milk survival. Using increasing concentrations of FeCl3, and the antibiotic streptonigrin we show that iron accumulates in the cytoplasm of ST4/74Δfur. We hypothesized that the accumulated iron could activate oxidative stress via Fenton's reaction leading to growth inhibition. However, the inhibition of ST4/74Δfur in milk was not due to Fenton's reaction, but due to the 'iron scarce' conditions of milk and microaerophilic incubation conditions which made the presence of the fur gene indispensable for Salmonella milk growth. Subsequently, survival studies of 14 other transcriptional mutants of ST4/74 in milk confirmed that RpoE-mediated response to extracytoplasmic stress is also important for the survival of Salmonella in milk. Though we have data only for fur and rpoE, many other Salmonella transcriptional factors could play important roles in the growth of Salmonella in milk, a theme for future research on Salmonella milk biology. Nevertheless, our data provide early insights into the biology of milk-associated Salmonella.
Collapse
Affiliation(s)
- Greeshma Bharathan
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | - Sunil Mundra
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates; Khalifa Centre for Genetic Engineering and Biotechnology, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Dania Mustafa Darwich
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | - Maitha Mohammad Saeed
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | - Ahad Saeed Ali Al Hafri
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | | | - Sajid Maqsood
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | - Priti Mudgil
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates
| | - Séamus Fanning
- UCD-Centre for Food Safety, Science Centre South, University College Dublin, Dublin, D04 N2E5, Ireland
| | - Shabarinath Srikumar
- Department of Food Science, College of Agriculture and Veterinary Medicine, UAE University, Al Ain, 15551, United Arab Emirates.
| |
Collapse
|
17
|
Suwandi A, Menon MB, Kotlyarov A, Grassl GA, Gaestel M. p38 MAPK/MK2 signaling stimulates host cells autophagy pathways to restrict Salmonella infection. Front Immunol 2023; 14:1245443. [PMID: 37771590 PMCID: PMC10523304 DOI: 10.3389/fimmu.2023.1245443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Autophagy plays an important role in recognizing and protecting cells from invading intracellular pathogens such as Salmonella. In this work, we investigated the role of p38MAPK/MK2 in modulating the host cell susceptibility to Salmonella infection. Inhibition of p38MAPK or MK2 led to a significant increase of bacterial counts in Salmonella infected mouse embryonic fibroblasts (MEFs), as well as in MK2-deficient (Mk2-/-) cells. Furthermore, western blot analysis showed that Mk2-/- cells have lower level of LC3 lipidation, which is the indicator of general autophagy compared to Mk2-rescued cells. In Mk2-/- cells, we also observed lower activated TANK-binding kinase-1 phosphorylation on Ser172 and p62/SQTM1-Ser403 phosphorylation, which are important to promote the translocation of p62 to ubiquitinated microbes and required for efficient autophagy of bacteria. Furthermore, immunofluorescence analysis revealed reduced colocalization of Salmonella with LC3 and p62 in MEFs. Inhibition of autophagy with bafilomycin A1 showed increased bacterial counts in treated cells compared to control cell. Overall, these results indicate that p38MAPK/MK2-mediated protein phosphorylation modulates the host cell susceptibility to Salmonella infection by affecting the autophagy pathways.
Collapse
Affiliation(s)
- Abdulhadi Suwandi
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Manoj B. Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Alexey Kotlyarov
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
18
|
Choi WS, Lee TH, Son SJ, Park YR, Lee TY, Javed A, Choe D, Kim SL, Kim SU, Lee SH. Antibacterial Effect of Berberrubine on the Salmonella typhimurium Type III Secretion System. Nat Prod Commun 2023; 18. [DOI: 10.1177/1934578x231189077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
The increasing antimicrobial drug resistance of Salmonella typhimurium has been a major public health problem in many countries. The type III secretion system (TTSS) of S typhimurium is an attractive target for the development of new antibiotics against this bacterium. In this study, we determined whether berberrubine derived from Berberis vulgaris could inhibit TTSS expression in S typhimurium . Berberrubine inhibited TTSS expression at a concentration of 60.9 µM. Additionally, TTSS-related mRNA expression of sipA, sipB, sopA, and sopD was downregulated in the presence of berberrubine. Collectively, we anticipate that berberrubine can be a useful tool to prevent the risks of infections caused by pathogenic bacteria.
Collapse
Affiliation(s)
- Won-Sik Choi
- Research Institute, JeonjinBio Co., Ltd, Daegu, The Republic of Korea
| | - Tae-Hun Lee
- Research Institute, JeonjinBio Co., Ltd, Daegu, The Republic of Korea
| | - Se-Jin Son
- Research Institute, JeonjinBio Co., Ltd, Daegu, The Republic of Korea
| | - Yeo Reum Park
- Research Institute, JeonjinBio Co., Ltd, Daegu, The Republic of Korea
| | - Tae-Young Lee
- Division of Emerging Infectious Disease & Vector Research, Center for Infectious Diseases Research, Osong, The Republic of Korea
| | - Ahsan Javed
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu, The Republic of Korea
| | - Deokyeong Choe
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu, The Republic of Korea
| | - Solomon L Kim
- California Northstate University College of Medicine, Elk Grove, CA, USA
| | - Sung Uk Kim
- Industrial Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, The Republic of Korea
- Department of Food Science and Engineering, Seowon University, Cheongju, The Republic of Korea
| | - Sang-Han Lee
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu, The Republic of Korea
- Food & Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu, The Republic of Korea
| |
Collapse
|
19
|
Ma Y, Fu W, Hong B, Wang X, Jiang S, Wang J. Antibacterial MccM as the Major Microcin in Escherichia coli Nissle 1917 against Pathogenic Enterobacteria. Int J Mol Sci 2023; 24:11688. [PMID: 37511446 PMCID: PMC10380612 DOI: 10.3390/ijms241411688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Probiotic Escherichia coli Nissle 1917 (EcN) possesses excellent antibacterial effects on pathogenic enterobacteria. The microcins MccM and MccH47 produced in EcN played critical roles, but they are understudied and poorly characterized, and the individual antibacterial mechanisms are still unclear. In this study, three EcN mutants (ΔmcmA, ΔmchB, and ΔmcmAΔmchB) were constructed and compared with wild-type EcN (EcN wt) to test for inhibitory effects on the growth of Escherichia coli O157: H7, Salmonella enterica (SE), and Salmonella typhimurium (ST). The antibacterial effects on O157: H7 were not affected by the knockout of mcmA (MccM) and mchB (MccH47) in EcN. However, the antibacterial effect on Salmonella declined sharply in EcN mutants ΔmcmA. The overexpressed mcmA gene in EcN::mcmA showed more efficient antibacterial activity on Salmonella than that of EcN wt. Furthermore, the EcN::mcmA strain significantly reduced the abilities of adhesion and invasion of Salmonella to intestinal epithelial cells, decreasing the invasion ability of ST by 56.31% (62.57 times more than that of EcN wt) while reducing the adhesion ability of ST by 50.14% (2.41 times more than that of EcN wt). In addition, the supernatant of EcN::mcmA culture significantly decreased the mRNA expression and secretion of IL-1β, TNF-α, and IL-6 on macrophages induced by LPS. The EcN::mcmA strain generated twice as much orange halo as EcN wt by CAS agar diffusion assay by producing more siderophores. MccM was more closely related to the activity of EcN against Salmonella, and MccM-overproducing EcN inhibited Salmonella growth by producing more siderophores-MccM to compete for iron, which was critical to pathogen growth. Based on the above, EcN::mcmA can be developed as engineered probiotics to fight against pathogenic enterobacteria colonization in the gut.
Collapse
Affiliation(s)
- Yi Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wei Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Bin Hong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xinfeng Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Shoujin Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jufang Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
20
|
Heyman O, Yehezkel D, Ciolli Mattioli C, Blumberger N, Rosenberg G, Solomon A, Hoffman D, Bossel Ben-Moshe N, Avraham R. Paired single-cell host profiling with multiplex-tagged bacterial mutants reveals intracellular virulence-immune networks. Proc Natl Acad Sci U S A 2023; 120:e2218812120. [PMID: 37399397 PMCID: PMC10334762 DOI: 10.1073/pnas.2218812120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/24/2023] [Indexed: 07/05/2023] Open
Abstract
Encounters between host cells and intracellular bacterial pathogens lead to complex phenotypes that determine the outcome of infection. Single-cell RNA sequencing (scRNA-seq) is increasingly used to study the host factors underlying diverse cellular phenotypes but has limited capacity to analyze the role of bacterial factors. Here, we developed scPAIR-seq, a single-cell approach to analyze infection with a pooled library of multiplex-tagged, barcoded bacterial mutants. Infected host cells and barcodes of intracellular bacterial mutants are both captured by scRNA-seq to functionally analyze mutant-dependent changes in host transcriptomes. We applied scPAIR-seq to macrophages infected with a library of Salmonella Typhimurium secretion system effector mutants. We analyzed redundancy between effectors and mutant-specific unique fingerprints and mapped the global virulence network of each individual effector by its impact on host immune pathways. ScPAIR-seq is a powerful tool to untangle bacterial virulence strategies and their complex interplay with host defense strategies that drive infection outcome.
Collapse
Affiliation(s)
- Ori Heyman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Dror Yehezkel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Camilla Ciolli Mattioli
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Neta Blumberger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Gili Rosenberg
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Aryeh Solomon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Dotan Hoffman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Noa Bossel Ben-Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
21
|
Dar MA, Bhat B, Nazir J, Saleem A, Manzoor T, Khan M, Haq Z, Bhat SS, Ahmad SM. Identification of SNPs Related to Salmonella Resistance in Chickens Using RNA-Seq and Integrated Bioinformatics Approach. Genes (Basel) 2023; 14:1283. [PMID: 37372463 DOI: 10.3390/genes14061283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Potential single nucleotide polymorphisms (SNPs) were detected between two chicken breeds (Kashmir favorella and broiler) using deep RNA sequencing. This was carried out to comprehend the coding area alterations, which cause variances in the immunological response to Salmonella infection. In the present study, we identified high impact SNPs from both chicken breeds in order to delineate different pathways that mediate disease resistant/susceptibility traits. Samples (liver and spleen) were collected from Salmonella resistant (K. favorella) and susceptible (broiler) chicken breeds. Salmonella resistance and susceptibility were checked by different pathological parameters post infection. To explore possible polymorphisms in genes linked with disease resistance, SNP identification analysis was performed utilizing RNA seq data from nine K. favorella and ten broiler chickens. A total of 1778 (1070 SNPs and 708 INDELs) and 1459 (859 SNPs and 600 INDELs) were found to be specific to K. favorella and broiler, respectively. Based on our results, we conclude that in broiler chickens the enriched pathways mostly included metabolic pathways like fatty acid metabolism, carbon metabolism and amino acid metabolism (Arginine and proline metabolism), while as in K. favorella genes with high impact SNPs were enriched in most of the immune-related pathways like MAPK signaling pathway, Wnt signaling pathway, NOD-like receptor signaling pathway, etc., which could be a possible resistance mechanism against salmonella infection. In K. favorella, protein-protein interaction analysis also shows some important hub nodes, which are important in providing defense against different infectious diseases. Phylogenomic analysis revealed that indigenous poultry breeds (resistant) are clearly separated from commercial breeds (susceptible). These findings will offer fresh perspectives on the genetic diversity in chicken breeds and will aid in the genomic selection of poultry birds.
Collapse
Affiliation(s)
- Mashooq Ahmad Dar
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences 3, 02-093 Warsaw, Poland
| | - Basharat Bhat
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
| | - Junaid Nazir
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
- Department of Clinical Biochemistry, Lovely Professional University, Phagwara 144402, India
| | - Afnan Saleem
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
| | - Tasaduq Manzoor
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
| | - Mahak Khan
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
| | - Zulfqarul Haq
- Indian Council of Medical Research Project, Division of Livestock Production and Management, F.V.Sc & AH, Shuhama, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar 190006, India
| | - Sahar Saleem Bhat
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar 190006, India
| | | |
Collapse
|
22
|
Xiong D, Song L, Chen Y, Jiao X, Pan Z. Salmonella Enteritidis activates inflammatory storm via SPI-1 and SPI-2 to promote intracellular proliferation and bacterial virulence. Front Cell Infect Microbiol 2023; 13:1158888. [PMID: 37325511 PMCID: PMC10266283 DOI: 10.3389/fcimb.2023.1158888] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
Salmonella Enteritidis is an important intracellular pathogen, which can cause gastroenteritis in humans and animals and threaten life and health. S. Enteritidis proliferates in host macrophages to establish systemic infection. In this study, we evaluated the effects of Salmonella pathogenicity island-1 (SPI-1) and SPI-2 to S. Enteritidis virulence in vitro and in vivo, as well as the host inflammatory pathways affected by SPI-1 and SPI-2. Our results show that S. Enteritidis SPI-1 and SPI-2 contributed to bacterial invasion and proliferation in RAW264.7 macrophages, and induced cytotoxicity and cellular apoptosis of these cells. S. Enteritidis infection induced multiple inflammatory responses, including mitogen-activated protein kinase (ERK-mediated) and Janus kinase-signal transducer and activator of transcript (STAT) (STAT2-mediated) pathways. Both SPI-1 and SPI-2 were necessary to induce robust inflammatory responses and ERK/STAT2 phosphorylation in macrophages. In a mouse infection model, both SPIs, especially SPI-2, resulted in significant production of inflammatory cytokines and various interferon-stimulated genes in the liver and spleen. Activation of the ERK- and STAT2-mediated cytokine storm was largely affected by SPI-2. S. Enteritidis ΔSPI-1-infected mice displayed moderate histopathological damage and drastically reduced bacterial loads in tissues, whereas only slight damage and no bacteria were observed in ΔSPI-2- and ΔSPI-1/SPI-2-infected mice. A survival assay showed that ΔSPI-1 mutant mice maintained a medium level of virulence, while SPI-2 plays a decisive role in bacterial virulence. Collectively, our findings indicate that both SPIs, especially SPI-2, profoundly contributed to S. Enteritidis intracellular localization and virulence by activating multiple inflammatory pathways.
Collapse
Affiliation(s)
- Dan Xiong
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Li Song
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Yushan Chen
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
Teklemariam AD, Al-Hindi RR, Albiheyri RS, Alharbi MG, Alghamdi MA, Filimban AAR, Al Mutiri AS, Al-Alyani AM, Alseghayer MS, Almaneea AM, Albar AH, Khormi MA, Bhunia AK. Human Salmonellosis: A Continuous Global Threat in the Farm-to-Fork Food Safety Continuum. Foods 2023; 12:foods12091756. [PMID: 37174295 PMCID: PMC10178548 DOI: 10.3390/foods12091756] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Salmonella is one of the most common zoonotic foodborne pathogens and a worldwide public health threat. Salmonella enterica is the most pathogenic among Salmonella species, comprising over 2500 serovars. It causes typhoid fever and gastroenteritis, and the serovars responsible for the later disease are known as non-typhoidal Salmonella (NTS). Salmonella transmission to humans happens along the farm-to-fork continuum via contaminated animal- and plant-derived foods, including poultry, eggs, fish, pork, beef, vegetables, fruits, nuts, and flour. Several virulence factors have been recognized to play a vital role in attaching, invading, and evading the host defense system. These factors include capsule, adhesion proteins, flagella, plasmids, and type III secretion systems that are encoded on the Salmonella pathogenicity islands. The increased global prevalence of NTS serovars in recent years indicates that the control approaches centered on alleviating the food animals' contamination along the food chain have been unsuccessful. Moreover, the emergence of antibiotic-resistant Salmonella variants suggests a potential food safety crisis. This review summarizes the current state of the knowledge on the nomenclature, microbiological features, virulence factors, and the mechanism of antimicrobial resistance of Salmonella. Furthermore, it provides insights into the pathogenesis and epidemiology of Salmonella infections. The recent outbreaks of salmonellosis reported in different clinical settings and geographical regions, including Africa, the Middle East and North Africa, Latin America, Europe, and the USA in the farm-to-fork continuum, are also highlighted.
Collapse
Affiliation(s)
- Addisu D Teklemariam
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rashad R Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed S Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mona G Alharbi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashail A Alghamdi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani A R Filimban
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdullah S Al Mutiri
- Laboratory Department, Saudi Food and Drug Authority, Riyadh 12843, Saudi Arabia
| | - Abdullah M Al-Alyani
- Laboratory Department, Saudi Food and Drug Authority, Jeddah 22311, Saudi Arabia
| | - Mazen S Alseghayer
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulaziz M Almaneea
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulgader H Albar
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Microbiology and Medical Parasitology, Faculty of Medicine, Jeddah University, Jeddah 23218, Saudi Arabia
| | - Mohsen A Khormi
- Department of Biological Sciences, Faculty of Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Interdisciplinary Life Science Program (PULSe), West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
24
|
Jeamsripong S, Kuldee M, Thaotumpitak V, Chuanchuen R. Antimicrobial resistance, Extended-Spectrum β-Lactamase production and virulence genes in Salmonella enterica and Escherichia coli isolates from estuarine environment. PLoS One 2023; 18:e0283359. [PMID: 37115770 PMCID: PMC10146452 DOI: 10.1371/journal.pone.0283359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/07/2023] [Indexed: 04/29/2023] Open
Abstract
The impact of antimicrobial resistance (AMR) on global public health has been widely documented. AMR in the environment poses a serious threat to both human and animal health but is frequently overlooked. This study aimed to characterize the association between phenotype and genotype of AMR, virulence genes and Extended-Spectrum β-Lactamase (ESBL) production from estuarine environment. The Salmonella (n = 126) and E. coli (n = 409) were isolated from oysters and estuarine water in Thailand. The isolates of Salmonella (96.9%) and E. coli (91.4%) showed resistance to at least one antimicrobial agent. Multidrug resistance (MDR) was 40.1% of Salmonella and 23.0% of E. coli. Resistance to sulfamethoxazole was most common in Salmonella (95.2%) and E. coli (77.8%). The common resistance genes found in Salmonella were sul3 (14.3%), followed by blaTEM (11.9%), and cmlA (11.9%), while most E. coli were blaTEM (31.5%) and tetA (25.4%). The ESBL production was detected in Salmonella (1.6%, n = 2) of which one isolate was positive to blaTEM-1. Eight E. coli isolates (2.0%) were ESBL producers, of which three isolates carried blaCTX-M-55 and one isolate was blaTEM-1. Predominant virulence genes identified in Salmonella were invA (77.0%), stn (77.0%), and fimA (69.0%), while those in E. coli isolates were stx1 (17.8%), lt (11.7%), and stx2 (1.2%). Logistic regression models showed the statistical association between resistance phenotype, virulence genes and ESBL production (p < 0.05). The findings highlighted that estuarine environment were potential hotspots of resistance. One Health should be implemented to prevent AMR bacteria spreading.
Collapse
Affiliation(s)
- Saharuetai Jeamsripong
- Research Unit in Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Mullika Kuldee
- Research Unit in Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Varangkana Thaotumpitak
- Research Unit in Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Rungtip Chuanchuen
- Research Unit in Microbial Food Safety and Antimicrobial Resistance, Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
25
|
Abstract
The ability of bacteria to respond to changes in their environment is critical to their survival, allowing them to withstand stress, form complex communities, and induce virulence responses during host infection. A remarkable feature of many of these bacterial responses is that they are often variable across individual cells, despite occurring in an isogenic population exposed to a homogeneous environmental change, a phenomenon known as phenotypic heterogeneity. Phenotypic heterogeneity can enable bet-hedging or division of labor strategies that allow bacteria to survive fluctuating conditions. Investigating the significance of phenotypic heterogeneity in environmental transitions requires dynamic, single-cell data. Technical advances in quantitative single-cell measurements, imaging, and microfluidics have led to a surge of publications on this topic. Here, we review recent discoveries on single-cell bacterial responses to environmental transitions of various origins and complexities, from simple diauxic shifts to community behaviors in biofilm formation to virulence regulation during infection. We describe how these studies firmly establish that this form of heterogeneity is prevalent and a conserved mechanism by which bacteria cope with fluctuating conditions. We end with an outline of current challenges and future directions for the field. While it remains challenging to predict how an individual bacterium will respond to a given environmental input, we anticipate that capturing the dynamics of the process will begin to resolve this and facilitate rational perturbation of environmental responses for therapeutic and bioengineering purposes.
Collapse
|
26
|
Shikov AE, Belousova ME, Belousov MV, Nizhnikov AA, Antonets KS. Salmonella-Based Biorodenticides: Past Applications and Current Contradictions. Int J Mol Sci 2022; 23:ijms232314595. [PMID: 36498920 PMCID: PMC9736839 DOI: 10.3390/ijms232314595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/03/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
The idea of using pathogens to control pests has existed since the end of the 19th century. Enterobacteria from the genus Salmonella, discovered at that time, are the causative agents of many serious diseases in mammals often leading to death. Mostly, the strains of Salmonella are able to infect a wide spectrum of hosts belonging to vertebrates, but some of them show host restriction. Several strains of these bacteria have been used as biorodenticides due to the host restriction until they were banned in many countries in the second part of the 20th century. The main reason for the ban was their potential pathogenicity for some domestic animals and poultry and the outbreaks of gastroenteritis in humans. Since that time, a lot of data regarding the host specificity and host restriction of different strains of Salmonella have been accumulated, and the complexity of the molecular mechanisms affecting it has been uncovered. In this review, we summarize the data regarding the history of studying and application of Salmonella-based rodenticides, discuss molecular systems controlling the specificity of Salmonella interactions within its multicellular hosts at different stages of infection, and attempt to reconstruct the network of genes and their allelic variants which might affect the host-restriction mechanisms.
Collapse
Affiliation(s)
- Anton E. Shikov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Maria E. Belousova
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
| | - Mikhail V. Belousov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Anton A. Nizhnikov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Kirill S. Antonets
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Correspondence:
| |
Collapse
|
27
|
Two Additional Connections between the Transcriptional Programs Controlling Invasion and Intracellular Replication of Salmonella: HilD-SprB Positively Regulates phoP and slyA. J Bacteriol 2022; 204:e0020422. [PMID: 36214553 PMCID: PMC9664945 DOI: 10.1128/jb.00204-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonella virulence relies on the ability of this bacterium to invade the intestinal epithelium and to replicate inside macrophages, which are functions mainly encoded in Salmonella pathogenicity islands 1 and 2 (SPI-1 and SPI-2), respectively. Complex regulatory programs control the expression of SPI-1 and SPI-2 and functionally related genes, involving the integration of ancestral regulators and regulators that Salmonella has acquired during its evolution. Interestingly, some previous studies have revealed cross talk between the regulatory programs for SPI-1 and SPI-2. Here, we report two additional connections between the regulatory programs controlling the expression of genes for invasion and intracellular replication. Our results show that the acquired regulators HilD and SprB, both encoded in SPI-1, induce, in a cascade fashion, the expression of PhoP and SlyA, two ancestral regulators that activate the expression of SPI-2 and other genes required for intracellular replication. We provide evidence supporting that the regulation of phoP and slyA by HilD-SprB was adapted during the divergence of Salmonella from its closer species, Escherichia coli, with the acquisition of SPI-1 and thus the gain of HilD and SprB, as well as through cis-regulatory evolution of phoP and slyA. Therefore, our study further expands the knowledge about the intricate regulatory network controlling the expression of virulence genes in Salmonella. IMPORTANCE Bacteria have developed diverse regulatory mechanisms to control genetic expression, in the case of pathogenic bacteria, to induce the expression of virulence genes in particular niches during host infection. In Salmonella, an intricate regulatory network has been determined, which controls the spatiotemporal expression of the SPI-1 and SPI-2 gene clusters that mediate the invasion to and the replication inside host cells, respectively. In this study, we report two additional pathways of cross talk between the transcriptional programs for SPI-1 and SPI-2. Additionally, our results support that these additional regulatory pathways were adapted during the divergence of Salmonella from its closer species, Escherichia coli. This study further expands the knowledge about the mechanisms determining the Salmonella virulence.
Collapse
|
28
|
Gao Y, Chen H, Li W, Zhang Y, Luo J, Zhao L, Shi F, Ye G, He X, Xu Z, Zhu L, Tang H, Li Y. Chloroform extracts of Atractylodes chinensis inhibit the adhesion and invasion of Salmonella typhimurium. Biomed Pharmacother 2022; 154:113633. [PMID: 36063647 DOI: 10.1016/j.biopha.2022.113633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
There are 27 million cases of Salmonella Typhimurium (STM) reported worldwide annually, which have resulted in 217,000 deaths to date. Thus, there is an urgent requirement to develop novel antibacterial agents to target the multidrug-resistant strains of STM. We evaluated the inhibitory effect of the chloroform extracts of Atractylodes chinensis (Ac-CE) on the virulence of STM in vitro and develop it as a potential antibacterial agent. First, we determined the in vitro effects of Ac-CE on STM biofilm formation, and swimming, swarming, and adhesion to mucin. Further, we evaluated the effect of Ac-CE on the adhesion and invasion of STM at the gene level. Lastly, we evaluated the inhibitory effect of Ac-CE on STM infectivity at the cellular level. Ac-CE could attenuate both the adhesion and invasion abilities of STM in vitro. At the gene level, it could inhibit the expression of flagella, pilus, biofilm, SPI-1, and SPI-2 genes, which are related to the adhesion and invasion ability of STM in cells. Ac-CE significantly downregulated the expression of inflammatory cytokines and the TLR4/MyD88/NF-κB pathway in an STM infection cell model. It also significantly recovered the expression of intestinal barrier-related genes and proteins in intestinal cells that are damaged during STM infection. Ac-CE is effective as an antivirulence agent in alleviating STM infection. Although the main components of Ac-CE were analyzed.We have not demonstrated the antivirulence effect of the active ingredients in Ac-CE. And the antivirulence effect of Ac-CE and its active ingredients warrant further in vivo studies.
Collapse
Affiliation(s)
- Yuanze Gao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Helin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Wen Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Yu Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Jie Luo
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, Guizhou, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Fei Shi
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Xiaoli He
- College of Science, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China.
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China.
| |
Collapse
|
29
|
Curcumin Stimulates the Overexpression of Virulence Factors in Salmonella enterica Serovar Typhimurium: In Vitro and Animal Model Studies. Antibiotics (Basel) 2022; 11:antibiotics11091230. [PMID: 36140009 PMCID: PMC9494991 DOI: 10.3390/antibiotics11091230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Salmonella spp. is one of the most common food poisoning pathogens and the main cause of diarrheal diseases in humans in developing countries. The increased Salmonella resistance to antimicrobials has led to the search for new alternatives, including natural compounds such as curcumin, which has already demonstrated a bactericidal effect; however, in Gram-negatives, there is much controversy about this effect, as it is highly variable. In this study, we aimed to verify the antibacterial activity of curcumin against the Salmonella enterica serovar Typhimurium growth rate, virulence, and pathogenicity. The strain was exposed to 110, 220 or 330 µg/mL curcumin, and by complementary methods (spectrophotometric, pour plate and MTT assays), we determined its antibacterial activity. To elucidate whether curcumin regulates the expression of virulence genes, Salmonella invA, fliC and siiE genes were investigated by quantitative real-time reverse transcription (qRT-PCR). Furthermore, to explore the effect of curcumin on the pathogenesis process in vivo, a Caenorhabditis elegans infection model was employed. No antibacterial activity was observed, even at higher concentrations of curcumin. All concentrations of curcumin caused overgrowth (35−69%) and increased the pathogenicity of the bacterial strain through the overexpression of virulence factors. The latter coincided with a significant reduction in both the lifespan and survival time of C. elegans when fed with curcumin-treated bacteria. Our data provide relevant information that may support the selective antibacterial effects of curcumin to reconsider the indiscriminate use of this phytochemical, especially in outbreaks of pathogenic Gram-negative bacteria.
Collapse
|
30
|
Dar MA, Ahmad SM, Bhat BA, Dar TA, Haq ZU, Wani BA, Shabir N, Kashoo ZA, Shah RA, Ganai NA, Heidari M. Comparative RNA-Seq analysis reveals insights in Salmonella disease resistance of chicken; and database development as resource for gene expression in poultry. Genomics 2022; 114:110475. [PMID: 36064074 DOI: 10.1016/j.ygeno.2022.110475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 07/07/2022] [Accepted: 07/24/2022] [Indexed: 11/04/2022]
Abstract
Salmonella, one of the major infectious diseases in poultry, causes considerable economic losses in terms of mortality and morbidity, especially in countries that lack effective vaccination programs. Besides being resistant to diseases, indigenous chicken breeds are also a potential source of animal protein in developing countries. For understanding the disease resistance, an indigenous chicken line Kashmir faverolla, and commercial broiler were selected. RNA-seq was performed after challenging the chicken with Salmonella Typhimurium. Comparative differential expression results showed that following infection, a total of 3153 genes and 1787 genes were differentially expressed in the liver and spleen, respectively. The genes that were differentially expressed included interleukins, cytokines, NOS2, Avβ-defensins, toll-like receptors, and other immune-related gene families. Most of the genes and signaling pathways involved in the innate and adaptive immune responses against bacterial infection were significantly enriched in the Kashmir faverolla. Pathway analysis revealed that most of the enriched pathways were MAPK signaling pathway, NOD-like receptor signaling pathway, TLR signaling pathway, PPAR signaling pathway, endocytosis, etc. Surprisingly some immune-related genes like TLRs were upregulated in the susceptible chicken breed. On postmortem examination, the resistant birds showed small lesions in the liver compared to large necrotic lesions in susceptible birds. The pathological manifestations and RNA sequencing results suggest a balancing link between resistance and infection tolerance in Kashmir faverolla. Here we also developed an online Poultry Infection Database (https://skuastk.org/pif/index.html), the first publicly available gene expression resource for disease resistance in chickens. The available database not only shows the data for gene expression in chicken tissues but also provides quick search, visualization and download capacity.
Collapse
Affiliation(s)
- Mashooq Ahmad Dar
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India; Department of Clinical Biochemistry/Biochemistry, University of Kashmir, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India.
| | - Basharat A Bhat
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry/Biochemistry, University of Kashmir, India
| | - Zulfqar Ul Haq
- Division of Livestock Poultry and Management, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Basharat A Wani
- Division of Veterinary Pathology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Nadeem Shabir
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Zahid Amin Kashoo
- Division of Veterinary Microbiology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Riaz Ahmad Shah
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | | | - Mohammad Heidari
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, 4279 E. Mount Hope Rd., East Lansing, MI 48823, USA
| |
Collapse
|
31
|
Demeter A, Jacomin AC, Gul L, Lister A, Lipscombe J, Invernizzi R, Branchu P, Macaulay I, Nezis IP, Kingsley RA, Korcsmaros T, Hautefort I. Computational prediction and experimental validation of Salmonella Typhimurium SopE-mediated fine-tuning of autophagy in intestinal epithelial cells. Front Cell Infect Microbiol 2022; 12:834895. [PMID: 36061866 PMCID: PMC9428466 DOI: 10.3389/fcimb.2022.834895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Macroautophagy is a ubiquitous homeostasis and health-promoting recycling process of eukaryotic cells, targeting misfolded proteins, damaged organelles and intracellular infectious agents. Some intracellular pathogens such as Salmonella enterica serovar Typhimurium hijack this process during pathogenesis. Here we investigate potential protein-protein interactions between host transcription factors and secreted effector proteins of Salmonella and their effect on host gene transcription. A systems-level analysis identified Salmonella effector proteins that had the potential to affect core autophagy gene regulation. The effect of a SPI-1 effector protein, SopE, that was predicted to interact with regulatory proteins of the autophagy process, was investigated to validate our approach. We then confirmed experimentally that SopE can directly bind to SP1, a host transcription factor, which modulates the expression of the autophagy gene MAP1LC3B. We also revealed that SopE might have a double role in the modulation of autophagy: Following initial increase of MAP1LC3B transcription triggered by Salmonella infection, subsequent decrease in MAP1LC3B transcription at 6h post-infection was SopE-dependent. SopE also played a role in modulation of the autophagy flux machinery, in particular MAP1LC3B and p62 autophagy proteins, depending on the level of autophagy already taking place. Upon typical infection of epithelial cells, the autophagic flux is increased. However, when autophagy was chemically induced prior to infection, SopE dampened the autophagic flux. The same was also observed when most of the intracellular Salmonella cells were not associated with the SCV (strain lacking sifA) regardless of the autophagy induction status before infection. We demonstrated how regulatory network analysis can be used to better characterise the impact of pathogenic effector proteins, in this case, Salmonella. This study complements previous work in which we had demonstrated that specific pathogen effectors can affect the autophagy process through direct interaction with autophagy proteins. Here we show that effector proteins can also influence the upstream regulation of the process. Such interdisciplinary studies can increase our understanding of the infection process and point out targets important in intestinal epithelial cell defense.
Collapse
Affiliation(s)
- Amanda Demeter
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- Department of Genetics, Eotvos Lorand University, Budapest, Hungary
| | | | - Lejla Gul
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - Ashleigh Lister
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - James Lipscombe
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - Rachele Invernizzi
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Priscilla Branchu
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Iain Macaulay
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
| | - Ioannis P. Nezis
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Robert A. Kingsley
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- *Correspondence: Tamas Korcsmaros,
| | | |
Collapse
|
32
|
Wang H, Tang Z, Xue B, Lu Q, Liu X, Zou Q. Salmonella Regulator STM0347 Mediates Flagellar Phase Variation via Hin Invertase. Int J Mol Sci 2022; 23:ijms23158481. [PMID: 35955615 PMCID: PMC9368917 DOI: 10.3390/ijms23158481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Salmonella enterica is one of the most important food-borne pathogens, whose motility and virulence are highly related to flagella. Flagella alternatively express two kinds of surface antigen flagellin, FliC and FljB, in a phenomenon known as flagellar phase variation. The molecular mechanisms by which the switching orientation of the Hin-composed DNA segment mediates the expression of the fljBA promoter have been thoroughly illustrated. However, the precise regulators that control DNA strand exchange are barely understood. In this study, we found that a putative response regulator, STM0347, contributed to the phase variation of flagellin in S. Typhimurium. With quantitative proteomics and secretome profiling, a lack of STM0347 was confirmed to induce the transformation of flagellin from FliC to FljB. Real-time PCR and in vitro incubation of SMT0347 with the hin DNA segment suggested that STM0347 disturbed Hin-catalyzed DNA reversion via hin degradation, and the overexpression of Hin was sufficient to elicit flagellin variation. Subsequently, the Δstm0347 strain was outcompeted by its parental strain in HeLa cell invasion. Collectively, our results reveal the crucial role of STM0347 in Salmonella virulence and flagellar phase variation and highlight the complexity of the regulatory network of Hin-modulated flagellum phase variation in Salmonella.
Collapse
Affiliation(s)
- Hongou Wang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.W.); (Z.T.); (Q.L.)
| | - Zhiheng Tang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.W.); (Z.T.); (Q.L.)
| | - Baoshuai Xue
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China;
| | - Qinghui Lu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.W.); (Z.T.); (Q.L.)
| | - Xiaoyun Liu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.W.); (Z.T.); (Q.L.)
- Correspondence: (X.L.); (Q.Z.); Tel.: +86-10-82805673 (X.L.); +86-10-8280-5070 (Q.Z.)
| | - Qinghua Zou
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (H.W.); (Z.T.); (Q.L.)
- Correspondence: (X.L.); (Q.Z.); Tel.: +86-10-82805673 (X.L.); +86-10-8280-5070 (Q.Z.)
| |
Collapse
|
33
|
A small molecule, C24H17ClN4O2S, inhibits the function of the type III secretion system in Salmonella Typhimurium. J Genet Eng Biotechnol 2022; 20:54. [PMID: 35380331 PMCID: PMC8982747 DOI: 10.1186/s43141-022-00336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/22/2022] [Indexed: 11/17/2022]
Abstract
Background Salmonella enterica serovar Typhimurium (S. Typhimurium) causes gastroenteritis and diarrhea in humans and food-producing animals. The type III secretion system (T3SS) has been known to be a potent virulence mechanism by injecting effector proteins into the cytosol of host cells. S. Typhimurium encodes two T3SSs by Salmonella pathogenicity islands 1 and 2. Previous studies showed that T3SS shared a potent virulence mechanism and molecular structure among several gram-negative bacteria. Therefore, T3SS has been identified as an attractive target in the development of novel therapeutics for the treatment of bacterial infections. Several studies reported that small-molecule compounds are able to inhibit functions of bacterial T3SSs. A small molecule, C24H17ClN4O2S, has been shown the ability to inhibit the activity of Yersinia pestis T3SS ATPase, YscN, resulting to block the secretion of effector proteins. In this study, we studied the effects and mechanism for SPI-1 T3SS inhibition of this compound in S. Typhimurium. Results We demonstrated that this compound prohibited the secretion of effector proteins from Salmonella via SPI-1 T3SS at 100 μM. As the result, bacterial invasion ability into epithelial cell cultures was reduced. In contrast with previous study, the C24H17ClN4O2S molecule did not inactivate the activity of SPI-1 T3SS ATPase, InvC, in Salmonella. However, we studied the global cellular effects of S. Typhimurium after being treated with this compound using a quantitative proteomic technique. These proteomic results showed that the main SPI-1 transcription regulator, InvF, and two effector proteins, SipA and SipC, were reduced in bacterial cells treated with the compound. Conclusions It may explain that action of the small-molecule compound, C24H17ClN4O2S, for blocking the secretion of SPI-1 T3SS in Salmonella is through inhibition of SPI-1 regulator, InvF, expression. Further studies are necessary to identify specific mechanisms for inhibition between this small-compound and InvF SPI-1 regulator protein.
Collapse
|
34
|
Xue J, Huang Y, Zhang H, Hu J, Pan X, Peng T, Lv J, Meng K, Li S. Arginine GlcNAcylation and Activity Regulation of PhoP by a Type III Secretion System Effector in Salmonella. Front Microbiol 2022; 12:825743. [PMID: 35126337 PMCID: PMC8811161 DOI: 10.3389/fmicb.2021.825743] [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] [Received: 11/30/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
Salmonella type III secretion system (T3SS) effector SseK3 is a glycosyltransferase delivered directly into the host cells to modify host protein substrates, thus manipulating host cellular signal transduction. Here, we identify and characterize the Arg-GlcNAcylation activity of SseK3 inside bacterial cells. Combining Arg-GlcNAc protein immunoprecipitation and mass spectrometry, we found that 60 bacterial proteins were GlcNAcylated during Salmonella infection, especially the two-component signal transduction system regulatory protein PhoP. Moreover, the Arg-GlcNAcylation of PhoP by SseK3 was detected in vivo and in vitro, and four arginine residues, Arg65, Arg66, Arg118, and Arg215 were identified as the GlcNAcylation sites. Site-directed mutagenesis showed that the PhoP R215A change significantly reduced the DNA-binding ability and arginine to alanine change at all four sites (PhoP 4RA) completely eliminated the DNA-binding ability, suggesting that Arg215 is essential for the DNA-binding activity of PhoP and GlcNAcylation of PhoP affects this activity. Additionally, GlcNAcylation of PhoP negatively regulated the activity of PhoP and decreased the expression of its downstream genes. Overall, our work provides an example of the intra-bacterial activities of the T3SS effectors and increases our understanding of endogenous Arg-GlcNAcylation.
Collapse
Affiliation(s)
- Juan Xue
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Yuxuan Huang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Hua Zhang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Jiaqingzi Hu
- Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Xing Pan
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Ting Peng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Jun Lv
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Kun Meng
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Shan Li
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Shan Li,
| |
Collapse
|
35
|
Wibisono FM, Faridah HD, Wibisono FJ, Tyasningsih W, Effendi MH, Witaningrum AM, Ugbo EN. Detection of invA virulence gene of multidrug-resistant Salmonella species isolated from the cloacal swab of broiler chickens in Blitar district, East Java, Indonesia. Vet World 2021; 14:3126-3131. [PMID: 35153402 PMCID: PMC8829414 DOI: 10.14202/vetworld.2021.3126-3131] [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: 06/23/2021] [Accepted: 11/02/2021] [Indexed: 11/28/2022] Open
Abstract
Background and Aim: The increasing number of multidrug-resistant (MDR) Salmonella species on poultry farms in Indonesia has caused concern regarding human health. This study was conducted to determine the presence of the virulence gene invA in MDR Salmonella species isolated from the cloacal swab of broiler chickens in Blitar district, East Java Province, Indonesia. Materials and Methods: Cloacal swab samples were collected by purposive sampling from 15 farms in four districts. Isolation and identification of bacteria were performed using standard microbiological techniques. Confirmation of MDR isolates was done using five different classes of antibiotics, including the beta-lactam, aminoglycoside, fluoroquinolone, phenicol, and monobactam groups. An antibiotic susceptibility test was conducted using the Kirby–Bauer disk diffusion method, and a polymerase chain reaction method was used to screen for the presence of invA. Results: It was observed that 32.26% (50/155) of the samples were positive for Salmonella species. Of these 50 Salmonella isolates, 7 (14%) were identified as MDR strains. An important finding was the detection of invA in all the seven MDR Salmonella strains (100%) isolated from the cloacal swab of broiler chickens in Blitar district, East Java Province. Conclusion: Veterinarians have an extremely important role in monitoring the use of antibiotics in farm animals to mitigate the rapid spread of MDR organisms in our environment, which can otherwise cause serious economic losses and also public health issues.
Collapse
Affiliation(s)
- Freshindy Marissa Wibisono
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hayyun Durrotul Faridah
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, Indonesia
| | - Freshinta Jellia Wibisono
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Wijaya Kusuma University Surabaya, Surabaya, Indonesia
| | - Wiwiek Tyasningsih
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Mustofa Helmi Effendi
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Adiana Mutamsari Witaningrum
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | |
Collapse
|
36
|
Zhao X, Wei S, Tian Q, Peng W, Tao Y, Bo R, Liu M, Li J. Eugenol exposure in vitro inhibits the expressions of T3SS and TIF virulence genes in Salmonella Typhimurium and reduces its pathogenicity to chickens. Microb Pathog 2021; 162:105314. [PMID: 34838999 DOI: 10.1016/j.micpath.2021.105314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Salmonella enterica serovar Typhimurium (S. Typhimurium) is a common food-borne pathogen, which has the ability to infect a wide range of hosts. The increasing emergence of drug-resistant strains urgently requires new alternative therapies. Eugenol has been shown to be very effective against drug-resistant strains of Gram-negative and Gram-positive bacteria. The purpose of this study is to explore the effects of eugenol on the virulence factors and pathogenicity of S. Typhimurium. METHODS The antibacterial activity of eugenol was investigated via the changes of cell morphology, fimbriae related-genes and virulence factors of S. Typhimurium, then the pathogenicity of S. Typhimurium pretreated by eugenol to chickens was evaluated. RESULTS Susceptibility testing showed that eugenol possessed significant antimicrobial activity. Scanning electron microscope analysis showed eugenol treatment deformed the morphology with damaged fimbriae structure of S. Typhimurium. Real time PCR assay confirmed eugenol significantly down-regulated the expressions of virulence factors (hilA, hilD, sipA, sipC, spiC, misL) of Type III secretion system (T3SS) and adherence genes (fimA, fimH, fimD, fimY, fimZ, stm0551) of Type I fimbriae (TIF). Animal experiment proved that the pathogenicity of S. Typhimurium exposed by eugenol was reduced, which was evidenced by the higher survival rate, weight gains and organs indexes, the lower bacterial loads in organs. Meanwhile, the duodenal histopathological changes were mitigated, with a significantly decline in the expressions of TNF-α, IL-6 and IL-18. CONCLUSION In summary, eugenol pretreatment may alleviate the pathogenicity of the S. Typhimurium to chickens via wrecking the fimbriae and inhibiting the mRNA expressions of virulence factors and adhesion molecules. These data dedicated the potential mechanisms of eugenol against S. Typhimurium in vitro.
Collapse
Affiliation(s)
- Xin Zhao
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China
| | - SiMin Wei
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - QiMing Tian
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - WeiLong Peng
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Ya Tao
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - RuoNan Bo
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - MingJiang Liu
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - JinGui Li
- School of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China.
| |
Collapse
|
37
|
Lamprokostopoulou A, Römling U. Yin and Yang of Biofilm Formation and Cyclic di-GMP Signaling of the Gastrointestinal Pathogen Salmonella enterica Serovar Typhimurium. J Innate Immun 2021; 14:275-292. [PMID: 34775379 PMCID: PMC9275015 DOI: 10.1159/000519573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022] Open
Abstract
Within the last 60 years, microbiological research has challenged many dogmas such as bacteria being unicellular microorganisms directed by nutrient sources; these investigations produced new dogmas such as cyclic diguanylate monophosphate (cyclic di-GMP) second messenger signaling as a ubiquitous regulator of the fundamental sessility/motility lifestyle switch on the single-cell level. Successive investigations have not yet challenged this view; however, the complexity of cyclic di-GMP as an intracellular bacterial signal, and, less explored, as an extracellular signaling molecule in combination with the conformational flexibility of the molecule, provides endless opportunities for cross-kingdom interactions. Cyclic di-GMP-directed microbial biofilms commonly stimulate the immune system on a lower level, whereas host-sensed cyclic di-GMP broadly stimulates the innate and adaptive immune responses. Furthermore, while the intracellular second messenger cyclic di-GMP signaling promotes bacterial biofilm formation and chronic infections, oppositely, Salmonella Typhimurium cellulose biofilm inside immune cells is not endorsed. These observations only touch on the complexity of the interaction of biofilm microbial cells with its host. In this review, we describe the Yin and Yang interactive concepts of biofilm formation and cyclic di-GMP signaling using S. Typhimurium as an example.
Collapse
Affiliation(s)
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Schwan CL, Lomonaco S, Bastos LM, Cook PW, Maher J, Trinetta V, Bhullar M, Phebus RK, Gragg S, Kastner J, Vipham JL. Genotypic and Phenotypic Characterization of Antimicrobial Resistance Profiles in Non-typhoidal Salmonella enterica Strains Isolated From Cambodian Informal Markets. Front Microbiol 2021; 12:711472. [PMID: 34603240 PMCID: PMC8481621 DOI: 10.3389/fmicb.2021.711472] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/03/2022] Open
Abstract
Non-typhoidal Salmonella enterica is a pathogen of global importance, particularly in low and middle-income countries (LMICs). The presence of antimicrobial resistant (AMR) strains in market environments poses a serious health threat to consumers. In this study we identified and characterized the genotypic and phenotypic AMR profiles of 81 environmental S. enterica strains isolated from samples from informal markets in Cambodia in 2018–2019. AMR genotypes were retrieved from the NCBI Pathogen Detection website (https://www.ncbi.nlm.nih.gov/pathogens/) and using ResFinder (https://cge.cbs.dtu.dk/services/) Salmonella pathogenicity islands (SPIs) were identified with SPIFinder (https://cge.cbs.dtu.dk/services/). Susceptibility testing was performed by broth microdilution according to the Clinical and Laboratory Standards Institute (CLSI) standard guidelines M100-S22 using the National Antimicrobial Resistance Monitoring System (NARMS) Sensititre Gram Negative plate. A total of 17 unique AMR genes were detected in 53% (43/81) of the isolates, including those encoding tetracycline, beta-lactam, sulfonamide, quinolone, aminoglycoside, phenicol, and trimethoprim resistance. A total of 10 SPIs (SPI-1, 3–5, 8, 9, 12–14, and centisome 63 [C63PI]) were detected in 59 isolates. C63PI, an iron transport system in SPI-1, was observed in 56% of the isolates (n = 46). SPI-1, SPI-4, and SPI-9 were present in 13, 2, and 5% of the isolates, respectively. The most common phenotypic resistances were observed to tetracycline (47%; n = 38), ampicillin (37%; n = 30), streptomycin (20%; n = 16), chloramphenicol (17%; n = 14), and trimethoprim-sulfamethoxazole (16%; n = 13). This study contributes to understanding the AMR genes present in S. enterica isolates from informal markets in Cambodia, as well as support domestic epidemiological investigations of multidrug resistance (MDR) profiles.
Collapse
Affiliation(s)
- Carla L Schwan
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| | - Sara Lomonaco
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, United States
| | - Leonardo M Bastos
- Department of Agronomy, Kansas State University, Manhattan, KS, United States
| | - Peter W Cook
- Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joshua Maher
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| | - Valentina Trinetta
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| | - Manreet Bhullar
- Department of Horticulture and Natural Resources, Kansas State University, Olathe, KS, United States
| | - Randall K Phebus
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| | - Sara Gragg
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| | - Justin Kastner
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Jessie L Vipham
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
39
|
Sibinelli-Sousa S, de Araújo-Silva AL, Hespanhol JT, Bayer-Santos E. Revisiting the steps of Salmonella gut infection with a focus on antagonistic interbacterial interactions. FEBS J 2021; 289:4192-4211. [PMID: 34546626 DOI: 10.1111/febs.16211] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022]
Abstract
A commensal microbial community is established in the mammalian gut during its development, and these organisms protect the host against pathogenic invaders. The hallmark of noninvasive Salmonella gut infection is the induction of inflammation via effector proteins secreted by the type III secretion system, which modulate host responses to create a new niche in which the pathogen can overcome the colonization resistance imposed by the microbiota. Several studies have shown that endogenous microbes are important to control Salmonella infection by competing for resources. However, there is limited information about antimicrobial mechanisms used by commensals and pathogens during these in vivo disputes for niche control. This review aims to revisit the steps that Salmonella needs to overcome during gut colonization-before and after the induction of inflammation-to achieve an effective infection. We focus on a series of reported and hypothetical antagonistic interbacterial interactions in which both contact-independent and contact-dependent mechanisms might define the outcome of the infection.
Collapse
Affiliation(s)
| | | | - Julia Takuno Hespanhol
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Ethel Bayer-Santos
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| |
Collapse
|
40
|
Jiang C, Huang X, Yao J, Yu L, Wei F, Yang A. The role of membrane contact sites at the bacteria-host interface. Crit Rev Microbiol 2021; 48:270-282. [PMID: 34403642 DOI: 10.1080/1040841x.2021.1961678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Membrane contact sites (MCSs) refer to the areas of close proximity between heterologous membranes. A growing body of evidence indicates that MCSs are involved in important cellular functions, such as cellular material transfer, organelle biogenesis, and cell growth. Importantly, the study of MCSs at the bacteria-host interface is an emerging popular research topic. Intracellular bacterial pathogens have evolved a variety of fascinating strategies to interfere with MCSs by injecting effectors into infected host cells. Bacteria-containing vacuoles establish direct physical contact with organelles within the host, ensuring vacuolar membrane integrity and energy supply from host organelles and protecting the vacuoles from the host endocytic pathway and lysosomal degradation. An increasing number of bacterial effectors from various bacterial pathogens hijack components of host MCSs to form the vacuole-organelle MCSs for material exchange. MCS-related events have been identified as new mechanisms of microbial pathogenesis to greatly improve bacterial survival and replication within host cells. In this review, we will discuss the recent advances in MCSs at the bacteria-host interface, focussing on the roles of MCSs mediated by bacterial effectors in microbial pathogenesis.
Collapse
Affiliation(s)
- Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Huang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jia Yao
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lihua Yu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Fujing Wei
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
41
|
Schultz BM, Melo-Gonzalez F, Salazar GA, Porto BN, Riedel CA, Kalergis AM, Bueno SM. New Insights on the Early Interaction Between Typhoid and Non-typhoid Salmonella Serovars and the Host Cells. Front Microbiol 2021; 12:647044. [PMID: 34276584 PMCID: PMC8282409 DOI: 10.3389/fmicb.2021.647044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica is a common source of food and water-borne infections, causing a wide range of clinical ailments in both human and animal hosts. Immunity to Salmonella involves an interplay between different immune responses, which are rapidly initiated to control bacterial burden. However, Salmonella has developed several strategies to evade and modulate the host immune responses. In this sense, the main knowledge about the pathogenicity of this bacterium has been obtained by the study of mouse models with non-typhoidal serovars. However, this knowledge is not representative of all the pathologies caused by non-typhoidal serovars in the human. Here we review the most important features of typhoidal and non-typhoidal serovars and the diseases they cause in the human host, describing the virulence mechanisms used by these pathogens that have been identified in different models of infection.
Collapse
Affiliation(s)
- Bárbara M Schultz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara N Porto
- Laboratory of Clinical and Experimental Immunology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.,Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
42
|
Gupta A, Bansal M, Liyanage R, Upadhyay A, Rath N, Donoghue A, Sun X. Sodium butyrate modulates chicken macrophage proteins essential for Salmonella Enteritidis invasion. PLoS One 2021; 16:e0250296. [PMID: 33909627 PMCID: PMC8081216 DOI: 10.1371/journal.pone.0250296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/02/2021] [Indexed: 12/28/2022] Open
Abstract
Salmonella Enteritidis is an intracellular foodborne pathogen that has developed multiple mechanisms to alter poultry intestinal physiology and infect the gut. Short chain fatty acid butyrate is derived from microbiota metabolic activities, and it maintains gut homeostasis. There is limited understanding on the interaction between S. Enteritidis infection, butyrate, and host intestinal response. To fill this knowledge gap, chicken macrophages (also known as HTC cells) were infected with S. Enteritidis, treated with sodium butyrate, and proteomic analysis was performed. A growth curve assay was conducted to determine sub-inhibitory concentration (SIC, concentration that do not affect bacterial growth compared to control) of sodium butyrate against S. Enteritidis. HTC cells were infected with S. Enteritidis in the presence and absence of SIC of sodium butyrate. The proteins were extracted and analyzed by tandem mass spectrometry. Our results showed that the SIC was 45 mM. Notably, S. Enteritidis-infected HTC cells upregulated macrophage proteins involved in ATP synthesis through oxidative phosphorylation such as ATP synthase subunit alpha (ATP5A1), ATP synthase subunit d, mitochondrial (ATP5PD) and cellular apoptosis such as Cytochrome-c (CYC). Furthermore, sodium butyrate influenced S. Enteritidis-infected HTC cells by reducing the expression of macrophage proteins mediating actin cytoskeletal rearrangements such as WD repeat-containing protein-1 (WDR1), Alpha actinin-1 (ACTN1), Vinculin (VCL) and Protein disulfide isomerase (P4HB) and intracellular S. Enteritidis growth and replication such as V-type proton ATPase catalytic subunit A (ATPV1A). Interestingly, sodium butyrate increased the expression of infected HTC cell protein involving in bacterial killing such as Vimentin (VIM). In conclusion, sodium butyrate modulates the expression of HTC cell proteins essential for S. Enteritidis invasion.
Collapse
Affiliation(s)
- Anamika Gupta
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Mohit Bansal
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Rohana Liyanage
- Department of Chemistry, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Abhinav Upadhyay
- Department of Animal Science, University of Connecticut, Storrs, Connecticut, United States of America
| | - Narayan Rath
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, Arkansas, United States of America
| | - Annie Donoghue
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture-Agriculture Research Station, Fayetteville, Arkansas, United States of America
| | - Xiaolun Sun
- Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, United States of America
| |
Collapse
|
43
|
Dos Santos AMP, Ferrari RG, Panzenhagen P, Rodrigues GL, Conte-Junior CA. Virulence genes identification and characterization revealed the presence of the Yersinia High Pathogenicity Island (HPI) in Salmonella from Brazil. Gene 2021; 787:145646. [PMID: 33848574 DOI: 10.1016/j.gene.2021.145646] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/22/2021] [Accepted: 04/07/2021] [Indexed: 11/30/2022]
Abstract
Salmonella spp. is one of the major agents of foodborne disease worldwide, and its virulence genes are responsible for the main pathogenic mechanisms of this micro-organism. The whole-genome sequencing (WGS) of pathogens has become a lower-cost and more accessible genotyping tool providing many gene analysis possibilities. This study provided an in silico investigation of 129 virulence genes, including plasmidial and bacteriophage genes from Brazilian strains' public Salmonella genomes. The frequency analysis of the four most sequenced serovars and a temporal analysis over the past four decades was also performed. The NCBI sequence reads archive (SRA) database comprised 1077 Salmonella public whole-genome sequences of strains isolated in Brazil between 1968 and 2018. Among the 1077 genomes, 775 passed in Salmonella in silico Typing (SISTR) quality control, which also identified 41 different serovars in which the four most prevalent were S. Enteritidis, S. Typhimurium, S. Dublin, and S. Heidelberg. Among these, S. Heidelberg presented the most distinct virulence profile, besides presenting Yersinia High Pathogenicity Island (HPI), rare and first reported in Salmonella from Brazil. The genes mgtC, csgC, ssaI and ssaS were the most prevalent within the 775 genomes with more than 99% prevalence. On the other hand, the less frequent genes were astA, iucBCD, tptC and shdA, with less than 1% frequency. All of the plasmids and bacteriophages virulence genes presented a decreasing trend between the 2000 s and 2010 s decades, except for the phage gene grvA, which increased in this period. This study provides insights into Salmonella virulence genes distribution in Brazil using freely available bioinformatics tools. This approach could guide in vivo and in vitro studies besides being an interesting method for the investigation and surveillance of Salmonella virulence. Moreover, here we propose the genes mgtC, csgC, ssaI and ssaS as additional targets for PCR identification of Salmonella in Brazil due to their very high frequency in the studied genomes.
Collapse
Affiliation(s)
- Anamaria M P Dos Santos
- Molecular & Analytical Laboratory Center, Faculty of Veterinary, Department of Food Technology, Universidade Federal Fluminense, Niterói, Brazil; Chemistry Institute, Food Science Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Center for Food Analysis (NAL-LADETEC), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafaela G Ferrari
- Chemistry Institute, Food Science Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Department of Animal Science, College for Agricultural Sciences, Federal University of Paraiba (CCA/UFPB), Areia, PB, Brazil.
| | - Pedro Panzenhagen
- Molecular & Analytical Laboratory Center, Faculty of Veterinary, Department of Food Technology, Universidade Federal Fluminense, Niterói, Brazil; Chemistry Institute, Food Science Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Center for Food Analysis (NAL-LADETEC), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Grazielle L Rodrigues
- Chemistry Institute, Food Science Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Center for Food Analysis (NAL-LADETEC), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Conte-Junior
- Molecular & Analytical Laboratory Center, Faculty of Veterinary, Department of Food Technology, Universidade Federal Fluminense, Niterói, Brazil; Chemistry Institute, Food Science Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Center for Food Analysis (NAL-LADETEC), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Health Quality Control, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Peñaloza D, Acuña LG, Barros MJ, Núñez P, Montt F, Gil F, Fuentes JA, Calderón IL. The Small RNA RyhB Homologs from Salmonella Typhimurium Restrain the Intracellular Growth and Modulate the SPI-1 Gene Expression within RAW264.7 Macrophages. Microorganisms 2021; 9:microorganisms9030635. [PMID: 33803635 PMCID: PMC8002944 DOI: 10.3390/microorganisms9030635] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Growing evidence indicates that small noncoding RNAs (sRNAs) play important regulatory roles during bacterial infection. In Salmonella Typhimurium, several sRNAs are strongly up-regulated within macrophages, but little is known about their role during the infection process. Among these sRNAs, the well-characterized paralogs RyhB-1 and RyhB-2 are two regulators of gene expression mainly related with the response to iron availability. To investigate the role of the sRNAs RyhB-1 and RyhB-2 from S. Typhimurium in the infection of RAW264.7 macrophages, we analyzed several phenotypic traits from intracellular mutant strains lacking one and both sRNAs. Deletion of RyhB-1 and/or RyhB-2 resulted in increased intracellular survival and faster replication within macrophages. The bacterial metabolic status inside macrophages was also analyzed, revealing that all the mutant strains exhibited higher intracellular levels of ATP and lower NAD+/NADH ratios than the wild type. Expression analyses from bacteria infecting macrophages showed that RyhB-1 and RyhB-2 affect the intra-macrophage expression of bacterial genes associated with the Salmonella pathogenicity island 1 (SPI-1) and the type III secretion system (T3SS). With a two-plasmid system and compensatory mutations, we confirmed that RyhB-1 and RyhB-2 directly interact with the mRNAs of the invasion chaperone SicA and the regulatory protein RtsB. Altogether, these results indicate that the RyhB homologs contribute to the S. Typhimurium virulence modulation inside macrophages by reducing the intracellular growth and down-regulating the SPI-1 gene expression.
Collapse
Affiliation(s)
- Diego Peñaloza
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
| | - Lillian G. Acuña
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
| | - M. José Barros
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
| | - Paula Núñez
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
| | - Fernanda Montt
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
| | - Fernando Gil
- Microbiota-Host Interactions and Clostridia Research Group, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile
- ANID-Millennium Science Initiative Program-Millennium Nucleus in the Biology of the Intestinal Microbiota, 8370186 Santiago, Chile
- Correspondence: (F.G.); (J.A.F.); (I.L.C.); Tel.: +56-2-2770-3065 (F.G.); +56-2-2661-8373 (J.A.F.); +56-2-2770-3422 (I.L.C.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile
- Correspondence: (F.G.); (J.A.F.); (I.L.C.); Tel.: +56-2-2770-3065 (F.G.); +56-2-2661-8373 (J.A.F.); +56-2-2770-3422 (I.L.C.)
| | - Iván L. Calderón
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370186 Santiago, Chile; (D.P.); (L.G.A.); (M.J.B.); (P.N.); (F.M.)
- Correspondence: (F.G.); (J.A.F.); (I.L.C.); Tel.: +56-2-2770-3065 (F.G.); +56-2-2661-8373 (J.A.F.); +56-2-2770-3422 (I.L.C.)
| |
Collapse
|
45
|
Genomic investigation of antimicrobial resistance determinants and virulence factors in Salmonella enterica serovars isolated from contaminated food and human stool samples in Brazil. Int J Food Microbiol 2021; 343:109091. [PMID: 33639477 DOI: 10.1016/j.ijfoodmicro.2021.109091] [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: 06/22/2020] [Revised: 01/07/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022]
Abstract
This study investigated the antimicrobial resistance determinants, virulence factors and identified serovars in 37 Salmonella enterica strains isolated from human stool and contaminated foods linked to outbreaks that occurred in Brazil over 7 years using whole genome sequencing (WGS). Phylogenetic analysis of selected serovars (S. Typhimurium, S. Infantis, S. London, and S. Johannesburg) was performed. Ten distinct serovars were identified and, 51% of the tested strains (n = 19) showed disagreement with the previous conventional serotyping. The antimicrobial resistance (AMR) determinants or plasmids varied among the strains. Resistome analysis revealed the presence of resistance genes to aminoglycosides [aac (6')-laa, aph (3″)-lb, aph (6)-ld, aadA1 and aadA2], sulfonamides (sul1), trimethoprin (dfrA8), fosfomycin (fosA7) and tetracyclines (tetA, tetB, tetC), as well as point mutations in parC (T57S) and gyrA (S83F). Plasmidome showed the presence of IncHI2, IncHI2A, IncFIB (S), IncFII (S), IncI1 and p0111 plasmids. Eight Salmonella pathogenicity islands and up to 102 stress and/or virulence genes were identified in the evaluated genomes. Virulence genes of K88 fimbrial adhesin were first reported in S. enterica (S. Pomona, S. Bredeney and S. Mbandaka strains). pilW gene was first identified in S. Pomona. Phylogenetic analysis showed that some serovars circulated in Brazil for decades, primarily within the poultry production chain. Findings highlighted the virulence and AMR determinants in strains that may lead to recurring food outbreaks.
Collapse
|
46
|
Barbosa PDPM, Ruviaro AR, Martins IM, Macedo JA, LaPointe G, Macedo GA. Enzyme-assisted extraction of flavanones from citrus pomace: Obtention of natural compounds with anti-virulence and anti-adhesive effect against Salmonella enterica subsp. enterica serovar Typhimurium. Food Control 2021. [DOI: 10.1016/j.foodcont.2020.107525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
47
|
Lee M, Ryu M, Joo M, Seo YJ, Lee J, Kim HM, Shin E, Yeom JH, Kim YH, Bae J, Lee K. Endoribonuclease-mediated control of hns mRNA stability constitutes a key regulatory pathway for Salmonella Typhimurium pathogenicity island 1 expression. PLoS Pathog 2021; 17:e1009263. [PMID: 33524062 PMCID: PMC7877770 DOI: 10.1371/journal.ppat.1009263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/11/2021] [Accepted: 01/01/2021] [Indexed: 01/04/2023] Open
Abstract
Bacteria utilize endoribonuclease-mediated RNA processing and decay to rapidly adapt to environmental changes. Here, we report that the modulation of hns mRNA stability by the endoribonuclease RNase G plays a key role in Salmonella Typhimurium pathogenicity. We found that RNase G determines the half-life of hns mRNA by cleaving its 5′ untranslated region and that altering its cleavage sites by genome editing stabilizes hns mRNA, thus decreasing S. Typhimurium virulence in mice. Under anaerobic conditions, the FNR-mediated transcriptional repression of rnc encoding RNase III, which degrades rng mRNA, and simultaneous induction of rng transcription resulted in rapid hns mRNA degradation, leading to the derepression of genes involved in the Salmonella pathogenicity island 1 (SPI-1) type III secretion system (T3SS). Together, our findings show that RNase III and RNase G levels-mediated control of hns mRNA abundance acts as a regulatory pathway upstream of a complex feed-forward loop for SPI-1 expression. Recent studies have shown that pathogenic bacteria with ribonuclease mutations display attenuated virulence, impaired mobility, and reduced proliferation in host cells. However, the molecular mechanisms underlying ribonuclease-associated pathogenesis have not yet been characterised. Here, we provide strong experimental evidence that the coordinated modulation of endoribonuclease activity constitutes an additional regulatory layer upstream of a complex feed-forward loop controlling global regulatory systems in the Salmonella pathogenicity island 1 (SPI-1) type III secretion system (T3SS). In addition, we showed that this regulatory pathway plays a key role in the virulence of S. Typhimurium in the host. Thus, our study improves the understanding of the mechanisms through which bacterial pathogens sense the host environment and respond precisely by expressing gene products required for adaptation to that particular niche.
Collapse
Affiliation(s)
- Minho Lee
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Minkyung Ryu
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Minju Joo
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Jaejin Lee
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Hong-Man Kim
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Eunkyoung Shin
- Department of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Ji-Hyun Yeom
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
| | - Yong-Hak Kim
- Department of Microbiology, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
- * E-mail: (Y-HK); (JB); (KL)
| | - Jeehyeon Bae
- Department of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
- * E-mail: (Y-HK); (JB); (KL)
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Dongjak-Gu, Seoul, Republic of Korea
- * E-mail: (Y-HK); (JB); (KL)
| |
Collapse
|
48
|
Nikam PS, Kingston JJ, Belagal Motatis AK. Oral co-administration of bivalent protein r-BL with U-Omp19 elicits mucosal immune responses and reduces S. Typhimurium shedding in BALB/c mice. Immunol Lett 2021; 231:61-67. [PMID: 33460704 DOI: 10.1016/j.imlet.2021.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/15/2020] [Accepted: 01/11/2021] [Indexed: 01/18/2023]
Abstract
The increase in international food trade and travel has dramatically increased the global incidences of Salmonellosis. In the light of widespread resistance to frontline antibiotics, oral vaccines remain the most reliable alternative. In this study, the fusion protein, r-BL was rationally constructed by splicing the Salmonella Typhimurium sseB and ompL genes through G4S linker by over-lap extension PCR. The oral coadministration of r-BL with B. abortus U-Omp19 protein with known protease inhibitor activity resulted in significant increase of mucosal IgA titres to antilog 4.5051 (p < 0.0001) and 4.806 (p < 0.0001) in the fecal samples and intestinal washes respectively. Antibody isotyping of the intestinal washes demonstrated increase in mucosal IgM, IgG1 and IgG2a isotypes also and demonstrated a significant reduction in fecal shedding of S. Typhimurium in challenge study. The r-BL + U-Omp19 treated mice demonstrated a complete termination of Salmonella fecal shedding by the 12th day of challenge as compared to other study groups. In summary, the bivalent protein r-BL when administered with the mucosal adjuvant U-Omp19 was successful in triggering mucosal arm of the immune system which forms the first line of defence in combating the infections caused by the enteric pathogen like Salmonella.
Collapse
Affiliation(s)
- Pradnya Sukhadev Nikam
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| | - Joseph J Kingston
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| | - Anil Kumar Belagal Motatis
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| |
Collapse
|
49
|
Lv Q, Lv Y, Dou X, Wassy SL, Jia G, Wei L, Yu Q, Deng X, Zhang C, Wang J. Myricetin inhibits the type III secretion system of Salmonella enterica serovar typhimurium by downregulating the Salmonella pathogenic island I gene regulatory pathway. Microb Pathog 2021; 150:104695. [PMID: 33418000 DOI: 10.1016/j.micpath.2020.104695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/29/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Based on the in-depth study of type III secretion systems (T3SS) in pathogenic bacteria, approaches targeting T3SS have become new alternative strategies to combat drug-resistant bacterial infections. As an important food-borne pathogen, Salmonella enterica serovar Typhimurium (S. Typhimurium) injects effector proteins into host cells through the T3SS to disrupt cell signaling and host responses. In this study, myricetin was screened for its ability to block the translocation function of effector proteins (SipA/SipB) using cell biology and molecular biology methods. It exerted strong effects on inhibiting the expression of Salmonella pathogenicity island 1 (SPI-1)-associated effector proteins without affecting S. Typhimurium growth and thus prevented S. Typhimurium from invading HeLa cells and ultimately inhibited S. Typhimurium-mediated cell damage. In an animal experiment, myricetin comprehensively protected mice from death and pathological damage. A further analysis of the mechanism of action showed that myricetin interfered with the regulatory network of SPI-1-related genes, resulting in a significant decrease in the levels of key effector proteins, and thus inhibited T3SS-mediated virulence. In summary, this study provides a solution for clinical resistance to S. Typhimurium infection and potential candidate compounds. Myricetin, a potential T3SS inhibitor, possesses effective biological activity and exerts protective effects in vitro and in vivo. Myricetin will likely be developed as a novel type of antibiotic targeting S. Typhimurium infections in the future.
Collapse
Affiliation(s)
- Qianghua Lv
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuanzhi Lv
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xinyi Dou
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Soromou Lanan Wassy
- Higher Institute of Sciences and Veterinary Medicine (ISSMV) of Dalaba, B.P 09, Guinea
| | - Guobin Jia
- HeBei Yuan Zheng Pharmaceutical CO., LTD, Shijiazhuang, Hebei, China
| | - Lijuan Wei
- Hebei Veterinary Medicine Technology Innovation Center, Shijiazhuang, Hebei, China
| | - Qinlei Yu
- Jilin Provincial Animal Disease Control Center, 4510 Xi'an Road, Changchun, China
| | - Xuming Deng
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chuanjin Zhang
- Shandong Veterinary Drug Quality Inspection Institute, Jinan, China.
| | - Jianfeng Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
50
|
Kommnick C, Hensel M. Correlative Light and Scanning Electron Microscopy to Study Interactions of Salmonella enterica with Polarized Epithelial Cell Monolayers. Methods Mol Biol 2021; 2182:103-115. [PMID: 32894490 DOI: 10.1007/978-1-0716-0791-6_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Live cell fluorescence imaging is the method of choice to visualize dynamic cellular processes in time and space, such as adhesion to and invasion of polarized epithelial cells by Salmonella enterica sv. Typhimurium. Scanning electron microscopy provides highest resolution of surface structures of infected cells, providing ultrastructure of the apical side of host cells and infecting Salmonella. Combining both methods toward correlative light and scanning electron microscopy (CLSEM) enables new insights in adhesion and invasion mechanisms regarding dynamics over time, and high spatial resolution with precise time lines. To correlate fast live cell imaging of polarized monolayer cells with scanning electron microscopy, we developed a robust method by using gold mesh grids as convenient CLSEM carriers for standard microscopes. By this, we were able to unravel the morphology of the apical structures of monolayers of polarized epithelial cells at distinct time points during Salmonella infection.
Collapse
Affiliation(s)
- Carina Kommnick
- Abteilung Mikrobiologie, CellNanOs-Center of Cellular Nanoanalytics Osnabrück, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Abteilung Mikrobiologie, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany.
- CellNanOs - Center of Cellular Nanoanalytics, Universität Osnabrück, Osnabrück, Germany.
| |
Collapse
|