1
|
Zhao H, Zhang X, Zhang N, Zhu L, Lian H. The interplay between Salmonella and host: Mechanisms and strategies for bacterial survival. CELL INSIGHT 2025; 4:100237. [PMID: 40177681 PMCID: PMC11964643 DOI: 10.1016/j.cellin.2025.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 04/05/2025]
Abstract
Salmonella, an intracellular pathogen, infects both humans and animals, causing diverse diseases such as gastroenteritis and enteric fever. The Salmonella type III secretion system (T3SS), encoded within its pathogenicity islands (SPIs), is critical for bacterial virulence by directly delivering multiple effectors into eukaryotic host cells. Salmonella utilizes these effectors to facilitate its survival and replication within the host through modulating cytoskeletal dynamics, inflammatory responses, the biogenesis of Salmonella-containing vacuole (SCV), and host cell survival. Moreover, these effectors also interfere with immune responses via inhibiting innate immunity or antigen presentation. In this review, we summarize the current progress in the survival strategies employed by Salmonella and the molecular mechanisms underlying its interactions with the host. Understanding the interplay between Salmonella and host can enhance our knowledge of the bacterium's pathogenic processes and provide new insights into how it manipulates host cellular physiological activities to ensure its survival.
Collapse
Affiliation(s)
- Hongyu Zhao
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Xinyue Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| | - Ningning Zhang
- Yale Stem Cell Center, New Haven, CT, 06520, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
- Yale Cooperative Center of Excellence in Hematology, New Haven, CT, 12208, USA
| | - Li Zhu
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Huan Lian
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
2
|
Girón-Pérez DA, Ley-Arteaga LV, Covantes-Rosales CE, Toledo-Ibarra GA, Díaz-Resendiz KJG, Bueno-Durán AY, Benitez-Trinidad AB, Navidad-Murrieta MS, Girón-Pérez MI. Differential infection dynamics in mononuclear and polymorphonuclear cells during Salmonella Typhimurium infection and in vitro exposure to diazoxon. Microb Pathog 2025; 200:107341. [PMID: 39884477 DOI: 10.1016/j.micpath.2025.107341] [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: 08/12/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
S. Typhimurium bacteria cause one of the most recurrent gastrointestinal diseases worldwide. This bacterium can settle in the gastrointestinal tract and internalize into different cellular strains, causing the formation of cellular reservoirs that subsequently lead to systemic dissemination. Exogenous factors such as pesticide exposure can also cause immunological alterations, increasing susceptibility to bacterial infection. The present work evaluated the infection capacity of Salmonella Typhimurium, during a short period (1 h) on mononuclear and polymorphonuclear cells previously exposed to diazoxon (1 h, during 4 h). Mononuclear cells were infected more frequently and in greater magnitude than polymorphonuclear cells. However, when actin polymerization and the release of reactive oxygen species (ROS) were analyzed, polymorphonuclear cells showed increased activity. These processes were evidenced by conformational changes during infection. This suggests differential dynamics of S. Typhimurium infection in mononuclear and polymorphonuclear cells previously exposed to diazoxon.
Collapse
Affiliation(s)
- Daniel Alberto Girón-Pérez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico.
| | - Leslie Verónica Ley-Arteaga
- Universidad Tecnológica de Nayarit, Carretera México 200, Km 9 63786, Col, 24 de febrero, Xalisco, 63786, Nayarit, Mexico
| | - Carlos Eduardo Covantes-Rosales
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Gladys Alejandra Toledo-Ibarra
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Karina Janice Guadalupe Díaz-Resendiz
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Adela Yolanda Bueno-Durán
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Alma Betsaida Benitez-Trinidad
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Migdalia Sarahy Navidad-Murrieta
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico
| | - Manuel Iván Girón-Pérez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, 63173, Nayarit, Mexico; Licenciatura en Biomedicine Ambiental Traslacional, Universidad Autónoma de Nayarit, Circuito C. Ney M. González, Ciudad del conocimiento, 63173, Tepic, Nay, Mexico.
| |
Collapse
|
3
|
Zhang H, Zhou G, Yang C, Nychas GJE, Zhang Y, Mao Y. The prevalence, distribution, and diversity of Salmonella isolated from pork slaughtering processors and retail outlets in the Shandong Province of China. Meat Sci 2025; 221:109734. [PMID: 39731976 DOI: 10.1016/j.meatsci.2024.109734] [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: 08/25/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
Salmonella is a foodborne pathogen of global significance and is highly prevalent in pork. This study investigated the prevalence, contamination distribution, virulence genes and antibiotic resistance of Salmonella in 3 pork processors in the Shandong Province of China. Samples were collected from 13 different sampling sources across the slaughter procedures (600 samples) as well as at retail outlets supplied by these processors (45 samples). The prevalence was 18.9 % among all the samples, with the highest prevalence observed in feces (40.0 %), lairage pens (38.0 %), and hides (34.0 %). A total of 6 serotypes were identified, with S. Rissen (46.3 %) and S. typhimurium (32.0 %) found to be the most prevalent serotypes. 86.8 % of Salmonella isolates were multi-drug resistant, with the majority of strains resistant to erythromycin, sulfisoxazole, and ampicillin. The multilocus sequence typing analysis revealed 6 STs were obtained from 45 isolates, with the dominant type ST469 accounting for 40.0 % of the total, which suggested a high possibility of cross-contamination between the plant processing chain and retail outlets. This work reveals the prevalence and correlation of Salmonella isolates between pork slaughter and retail outlets and acts as a case-study for stakeholders wishing to improve pork supply chain hygiene, control cross-contamination between the various slaughtering processes, and obtain continuous updates on Salmonella surveillance.
Collapse
Affiliation(s)
- Haoqi Zhang
- Laboratory of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, PR China; National R&D Center for Beef Processing Technology, Tai'an, Shandong 271018, PR China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China.
| | - Guanghui Zhou
- Laboratory of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, PR China; National R&D Center for Beef Processing Technology, Tai'an, Shandong 271018, PR China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China
| | - Caishuai Yang
- Laboratory of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, PR China; National R&D Center for Beef Processing Technology, Tai'an, Shandong 271018, PR China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China
| | - George-John E Nychas
- International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China; Laboratory of Microbiology and Biotechnology of Foods, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, Iera Odos 75, Athens 11855, Greece
| | - Yimin Zhang
- Laboratory of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, PR China; National R&D Center for Beef Processing Technology, Tai'an, Shandong 271018, PR China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China.
| | - Yanwei Mao
- Laboratory of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, PR China; National R&D Center for Beef Processing Technology, Tai'an, Shandong 271018, PR China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, PR China.
| |
Collapse
|
4
|
Xu L, Wang J, Zhong J, Lin W, Shen G, He N, Mao X, Fu C, Huang Z, Zhao F, Ye X, Zhu Y, Zheng M, Li H, Wang LL, Zhong K, Zhu L, Chen YY. Multiomics identify the gene expression signature of the spinal cord during aging process. Commun Biol 2025; 8:193. [PMID: 39920442 PMCID: PMC11806003 DOI: 10.1038/s42003-025-07475-4] [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: 06/15/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Age-related long-term disability is attracting increasing attention due to the growing ageing population worldwide. However, the current understanding of the senescent spinal cord remains insufficient. Bulk RNA sequencing reveals that 526 genes are upregulated and 300 genes are downregulated in senescent spinal cords. Pathway enrichment analysis of differentially expressed genes shows that senescence in spinal cords is related to phagosome function, neuroinflammation, ferroptosis, and necroptosis. Prediction of upstream transcription factors and interactome analysis identify Spi1 as a transcription factor that potentially plays a core role in senescent spinal cords. Spatial transcriptomics illustrates the spatial distribution of the transcriptomic landscape in both young and senescent spinal cords and identifies distinct neuronal and glial subtypes. The ferroptosis-associated gene Fth1 is upregulated in aged spinal cords. Flow cytometry reveals increased accumulation of free Fe2+ and ROS in senescent mixed glial cells; however, CCK-8 assays reveal that these cells are resistant to ferroptosis. SiRNA and lentivirus experiments indicate that the overexpression of Fth1 in normal mixed glial cells reduces their sensitivity to ferroptosis, whereas Fth1 knockdown increases their sensitivity to ferroptosis. In summary, spatial and bulk transcriptomics elucidate the transcriptional characteristics of young versus senescent spinal cords, thus highlighting the role of Fth1 in mediating ferroptosis resistance in senescent mixed glial cells.
Collapse
Affiliation(s)
- Lintao Xu
- Department of Obstetrics of the Second Affiliated Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyu Wang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- Department of Obstetrics of the Second Affiliated Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Lin
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Gerong Shen
- Department of Orthopaedics of Sir Run Run Shaw Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning He
- Department of Orthopaedics of Sir Run Run Shaw Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingjia Mao
- Department of Orthopaedics of Sir Run Run Shaw Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunyan Fu
- Department of Obstetrics of the Second Affiliated Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaobo Huang
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengdong Zhao
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Ye
- Department of Neurosurgery, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yongjian Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mingzhi Zheng
- Department of Pharmacology, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hui Li
- Laboratory of Animal Research Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin-Lin Wang
- Department of Orthopaedics of Sir Run Run Shaw Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China.
| | - Kai Zhong
- Department of Pharmacology, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China.
| | - Lijun Zhu
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Ying-Ying Chen
- Department of Obstetrics of the Second Affiliated Hospital and Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Torsilieri HM, Upchurch CM, Leitinger N, Casanova JE. Salmonella-induced cholesterol accumulation in infected macrophages suppresses autophagy via mTORC1 activation. Mol Biol Cell 2025; 36:ar3. [PMID: 39602284 PMCID: PMC11742112 DOI: 10.1091/mbc.e24-06-0283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/07/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative bacillus that infects the host intestinal epithelium and resident macrophages. Many intracellular pathogens induce an autophagic response in host cells but have evolved mechanisms to subvert that response. Autophagy is closely linked to cellular cholesterol levels; mTORC1 senses increased cholesterol in lysosomal membranes, leading to its hyperactivity and suppression of autophagy. Previous studies indicate that Salmonella infection induces dramatic accumulation of cholesterol in macrophages, a fraction of which localizes to Salmonella containing vacuoles (SCVs). We previously reported that the bacterial effector protein SseJ triggers cholesterol accumulation through a signaling cascade involving focal adhesion kinase (FAK) and Akt. Here we show that mTORC1 is recruited to SCVs and is hyperactivated in a cholesterol-dependent manner. If cholesterol accumulation is prevented pharmacologically or through mutation of sseJ, autophagy is induced and bacterial survival is attenuated. Notably, the host lipid transfer protein OSBP (oxysterol binding protein 1) is also recruited to SCVs and its activity is necessary for both cholesterol transfer to SCVs and mTORC1 activation during infection. Finally, lipidomic analysis of Salmonella-infected macrophages revealed new insights into how Salmonella may manipulate lipid homeostasis to benefit its survival. We propose that S. Typhimurium induces cholesterol accumulation through SseJ to activate mTORC1, preventing autophagic clearance of bacteria.
Collapse
Affiliation(s)
- Holly M. Torsilieri
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22903
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - James E. Casanova
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22903
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22903
| |
Collapse
|
6
|
Cui X, Wang Y, Liu Z, Zhao M, Zhu M, Yu W, Lu B, Xu H, Liu J, Liao N, Shi J, Peng D, Niu S, Shen J, Qiu J, Yu L. Ginsenoside Ro improves Salmonella Typhimurium-induced colitis through inhibition of the virulence factors SopB and SopE2 via the RAC1/CDC42/ARP2/3 pathway. FASEB J 2024; 38:e70282. [PMID: 39734277 PMCID: PMC11695707 DOI: 10.1096/fj.202401712r] [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: 07/25/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) poses a serious threat to human and animal health, and there is an urgent need to develop new therapeutic agents. In our in vivo study, ginsenoside Ro (Ro) reduced the mortality rate of S. Typhimurium-infected mice by effectively improving three key disease activity index (DAI) indicators. In particular, ginsenoside Ro inhibited S. Typhimurium-induced colitis by reversing colon length shortening; alleviating pathological damage to the colon; decreasing the levels of IL-1β, TNF-α, IFN-γ, and IL-6; and decreasing the activities of MPO and EPO, while increasing the levels of IL-10 as well as the colon epithelial barrier and tight junction-related genes (Mucin 1, Mucin 2, Occludin, Claudin-3, and ZO-1). Furthermore, ginsenoside Ro reduced CFUs in the liver, spleen, colon, and feces. In a mechanistic in vitro study, ginsenoside Ro reduced CFUs in HeLa and Raw264.7 cells, which was associated with ginsenoside Ro inhibition of the recruited S. Typhimurium-containing vacuole (SCV) biomarkers LC3, Rab7, GAL8, and NDP52. Molecular docking results revealed that the binding energies of ginsenoside Ro and SopB and ginsenoside Ro and SopE2 were as high as -11.3 and -9.7 kcal/mol, respectively, as verified by CETSA and DARTS assays. Moreover, ginsenoside Ro at 100 and 200 μM significantly decreased the enzyme activities and expression of SopB and SopE2. Finally, ginsenoside Ro inhibited the membrane ruffling caused by SopB-regulated Arf6/Cyth2/Arf1-, RAC1-, and CDC42-driven Arp2/3-dependent actin polymerization and the SopE2-regulated CDC42/Arp2/3 signaling pathway. In summary, our findings suggest that ginsenoside Ro is a potential lead compound for therapeutic use against S. Typhimurium infection, and these findings lay a foundation for its further development.
Collapse
Affiliation(s)
- Xinhua Cui
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Yang Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Ziyan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Meng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Mingmei Zhu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Wanlu Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Baochun Lu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Hongyue Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Jiajia Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Ning Liao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Jinyang Shi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Danping Peng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Sijia Niu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Jiayi Shen
- Key Laboratory of Organo‐Pharmaceutical Chemistry of Jiangxi ProvinceGannan Normal UniversityGanzhouChina
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| | - Lu Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious DiseasesFirst Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
7
|
Han J, Aljahdali N, Zhao S, Tang H, Harbottle H, Hoffmann M, Frye JG, Foley SL. Infection biology of Salmonella enterica. EcoSal Plus 2024; 12:eesp00012023. [PMID: 38415623 PMCID: PMC11636313 DOI: 10.1128/ecosalplus.esp-0001-2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/31/2023] [Indexed: 02/29/2024]
Abstract
Salmonella enterica is the leading cause of bacterial foodborne illness in the USA, with an estimated 95% of salmonellosis cases due to the consumption of contaminated food products. Salmonella can cause several different disease syndromes, with the most common being gastroenteritis, followed by bacteremia and typhoid fever. Among the over 2,600 currently identified serotypes/serovars, some are mostly host-restricted and host-adapted, while the majority of serotypes can infect a broader range of host species and are associated with causing both livestock and human disease. Salmonella serotypes and strains within serovars can vary considerably in the severity of disease that may result from infection, with some serovars that are more highly associated with invasive disease in humans, while others predominantly cause mild gastroenteritis. These observed clinical differences may be caused by the genetic make-up and diversity of the serovars. Salmonella virulence systems are very complex containing several virulence-associated genes with different functions that contribute to its pathogenicity. The different clinical syndromes are associated with unique groups of virulence genes, and strains often differ in the array of virulence traits they display. On the chromosome, virulence genes are often clustered in regions known as Salmonella pathogenicity islands (SPIs), which are scattered throughout different Salmonella genomes and encode factors essential for adhesion, invasion, survival, and replication within the host. Plasmids can also carry various genes that contribute to Salmonella pathogenicity. For example, strains from several serovars associated with significant human disease, including Choleraesuis, Dublin, Enteritidis, Newport, and Typhimurium, can carry virulence plasmids with genes contributing to attachment, immune system evasion, and other roles. The goal of this comprehensive review is to provide key information on the Salmonella virulence, including the contributions of genes encoded in SPIs and plasmids during Salmonella pathogenesis.
Collapse
Affiliation(s)
- Jing Han
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Nesreen Aljahdali
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
- Biological Science Department, College of Science, King Abdul-Aziz University, Jeddah, Saudi Arabia
| | - Shaohua Zhao
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Hailin Tang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Heather Harbottle
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Maria Hoffmann
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland, USA
| | - Jonathan G. Frye
- Agricutlutral Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Steven L. Foley
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
8
|
Zhang K, Wang P, Li S, Xie X, Wang Z, Li Y, Jiao X, Li Q. Type I-E CRISPR-Cas system regulates fimZY and T3SS1 genes expression in Salmonella enterica serovar Pullorum. Vet Microbiol 2024; 299:110301. [PMID: 39561528 DOI: 10.1016/j.vetmic.2024.110301] [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: 06/26/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024]
Abstract
Clustered regularly interspaced short palindromic repeats and associated Cas proteins (CRISPR-Cas) provide prokaryotes with adaptive immunity against invasion by plasmids or phages. In Salmonella, the type I-E CRISPR-Cas system is typically considered silent in immunity against foreign genetic elements. To elucidate the role of the CRISPR-Cas system, we chose Salmonella enterica serovar Pullorum S06004 as a model organism due to its four spacers and well-defined biological characteristics observed in previous studies. Western blot analysis revealed expression of Cas3 in S06004 cultured in vitro, but plasmid transformation assays demonstrated that both wild-type (WT) and S06004 strains overexpressing LeuO (a positive regulator of CRISPR-Cas) showed no immunity against the target plasmid. RNA-Seq analysis detected significant downregulation of the fim cluster, encoding type I fimbriae, and T3SS1-related genes in the cas cluster mutant compared to the WT. This downregulation was further confirmed in mutants of CR1 and individual cas genes by qRT-PCR. Consequently, mutants of CR1 and cas clusters exhibited decreased invasion of chicken hepatocellular carcinoma cells. The consistent regulation of T3SS1 genes by the CRISPR-Cas system in S. Pullorum, S. Enteritidis, and S. Typhimurium indicates a common role for the type I-E CRISPR-Cas system in promoting bacterial virulence. However, the specific molecular mechanisms underlying this regulation require further investigation.
Collapse
Affiliation(s)
- Kai Zhang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China
| | - Pengyu Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China
| | - Shanshan Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China
| | - Xiaolei Xie
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China
| | - Zhenyu Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China
| | - Yang Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China
| | - Qiuchun Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, China.
| |
Collapse
|
9
|
Abdel-Raheem SM, El-Hamid MIA, Khamis T, Baz HA, Omar AE, Gad WM, El-Azzouny MM, Habaka MAM, Mohamed RI, Elkenawy ME, Dawod RE, Elalfy EA, Ibrahim D. Comprehensive efficacy of nano-formulated mixed probiotics on broiler chickens' performance and Salmonella Typhimurium challenge. Poult Sci 2024; 103:104334. [PMID: 39366292 PMCID: PMC11489064 DOI: 10.1016/j.psj.2024.104334] [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: 05/22/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024] Open
Abstract
The increasing recognition of the potential advantages beyond nanoencapsulation of probiotics gained great attention owing to effective properties. Hence, we provided the most in-depth look into the influence of nanoformulated multi strain probiotics; BLB-NPs comprising Bacillus subtilis ATCC19659, Lactobacillus plantarum ATCC8014 and Bifidobacterium bifidum ATCC29521 on growth performance, antioxidant status and intestinal immunity supporting the defense against Salmonella Typhimurium (S. Typhimurium) challenge in broilers chickens. A total of 2,800 one-day-old male Ross 308 boiler chicks were divided into 7 groups; 1 control without additives, 3 probiotics [fed control diets mixed with B. subtilis, L. plantarum and B. bifidum (BLB) at concentrations of 1 × 104 (BLBI), 1 × 106 (BLBII) and 1 × 108 (BLBIII) CFU /kg diet, respectively] and 3 nanoencapsulated probiotics [fed control diets supplemented with BLB loaded nanoparticles (BLB-NPs) at concentrations of 1 × 104 (BLB-NPsI), 1 × 106 (BLB-NPsII) and 1 × 108 (BLB-NPsIII) CFU /kg diet, respectively]. All previous groups were challenged at d 22 of age with S. Typhimurium. Birds fed BLB-NPs II and III exhibited better weight gain and FCR simultaneously with upregulation in nutrients transporters genes (LAT-1, PepT-1, CAT-1 and SGLT1) even after S. Typhimurium challenge. Upregulation of immmune related genes (IL-1β, IL-6, IL-8, MyD88, NF-kB, CCL20, CXCLi2, TLR-2, TLR-4 and SOCS1) was prominently subsided in BLB-NPsIII fed group. The strengthening ability of BLB-NPs for broilers' intestinal barriers was evidenced by augmented expression of JAM, MUC-2, occludin and FABP-2 genes, diminished S. Typhimurium counts and suppressed its virulence related genes (HilA and SopD) with restored histopathological pictures of cecum. Notably, post dietary inclusion of higher levels of BLB-NPsIII, the abundance of beneficial Biofidobacterium and Lactobacillus species was dominated over harmful E. coli ones. Birds fortified with BLB-NPs displayed potent antioxidant potential signified by boosting serum and intestinal antioxidant markers alongside reducing oxidative ones. Overall, the abovementioned positive outcomes of BLB-NPs encouraged their potential application in poultry feed to attain superior performance and elicit protective immunity against S. Typhimurium infection.
Collapse
Affiliation(s)
- Sherief M Abdel-Raheem
- Department of Public Health, College of Veterinary Medicine, King Faisal University, Hofuf, Al-Ahsa 31982, Saudi Arabia; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Marwa I Abd El-Hamid
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Tarek Khamis
- Department of Pharmacology and Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Heba A Baz
- Veterinary Education Hospital, Poultry diseases, Fac2ulty of Veterinary Medicine, Zagazig, 44511, Egypt
| | - Anaam E Omar
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Egypt
| | - Wafaa M Gad
- Department of Bacteriology, Animal Health Research Institute (AHRI), Mansoura Branch Agriculture Research Center, Mansoura 35511, Egypt
| | - Mona M El-Azzouny
- Department of Bacteriology, Animal Health Research Institute (AHRI), Zagazig Branch, Agriculture Research Center (ARC), Zagazig 44516, Egypt
| | - Manal A M Habaka
- Department of Poultry and Rabbits Diseases, Animal Health Research Institute (AHRI), Zagazig Branch, Agriculture Research Center (ARC), Zagazig 44516, Egypt
| | - Rania I Mohamed
- Department of Pathology, Agricultural Research Center (ARC), Animal Health Research Institute, Mansoura Provincial Laboratory (AHRI-Mansoura), Mansoura, 35511, Egypt
| | - Mona E Elkenawy
- Department of Biochemistry, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), Mansoura, Egypt
| | - Rehab E Dawod
- Department of Bacteriology, Animal Health Research Institute (AHRI), Damietta Branch, Agriculture Research center (ARC), Damietta, Egypt
| | - Eman A Elalfy
- Department of Biochemistry, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), Mansoura, Egypt
| | - Doaa Ibrahim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Egypt
| |
Collapse
|
10
|
Shuster M, Lyu Z, Augenstreich J, Mathur S, Ganesh A, Ling J, Briken V. Salmonella Typhimurium infection inhibits macrophage IFNβ signaling in a TLR4-dependent manner. Infect Immun 2024; 92:e0009824. [PMID: 39269166 PMCID: PMC11475681 DOI: 10.1128/iai.00098-24] [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: 03/01/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Type I Interferons (IFNs) generally have a protective role during viral infections, but their function during bacterial infections is dependent on the bacterial species. Legionella pneumophila, Shigella sonnei and Mycobacterium tuberculosis can inhibit type I IFN signaling. Here we examined the role of type I IFN, specifically IFNβ, in the context of Salmonella enterica serovar Typhimurium (STm) macrophage infections and the capacity of STm to inhibit type I IFN signaling. We demonstrate that IFNβ has no effect on the intracellular growth of STm in infected bone marrow derived macrophages (BMDMs) derived from C57BL/6 mice. STm infection inhibits IFNβ signaling but not IFNγ signaling in a murine macrophage cell line. We show that this inhibition is independent of the type III and type VI secretion systems expressed by STm and is also independent of bacterial phagocytosis. The inhibition is Toll-like receptor 4 (TLR4)-dependent as the TLR4 ligand, lipopolysaccharide (LPS), alone is sufficient to inhibit IFNβ-mediated signaling. Cells downregulated their surface levels of IFNα/β receptor 1 (IFNAR1) in response to LPS, which may be mediating our observed inhibition. Lastly, we examined this inhibition in the context of TLR4-deficient BMDMs as well as TLR4 RNA interference and we observed a loss of inhibition with LPS stimulation as well as STm infection. In summary, we show that macrophages exposed to STm have reduced IFNβ signaling via crosstalk with TLR4 signaling, which may be mediated by reduced host cell surface IFNAR1, and that IFNβ signaling does not affect cell-autonomous host defense against STm.
Collapse
Affiliation(s)
- Michael Shuster
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Shrestha Mathur
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Akshaya Ganesh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
11
|
Sujith S, Naresh R, Srivisanth BU, Sajeevan A, Rajaramon S, David H, Solomon AP. Aptamers: precision tools for diagnosing and treating infectious diseases. Front Cell Infect Microbiol 2024; 14:1402932. [PMID: 39386170 PMCID: PMC11461471 DOI: 10.3389/fcimb.2024.1402932] [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: 03/18/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Infectious diseases represent a significant global health challenge, with bacteria, fungi, viruses, and parasitic protozoa being significant causative agents. The shared symptoms among diseases and the emergence of new pathogen variations make diagnosis and treatment complex. Conventional diagnostic methods are laborious and intricate, underscoring the need for rapid, accurate techniques. Aptamer-based technologies offer a promising solution, as they are cost-effective, sensitive, specific, and convenient for molecular disease diagnosis. Aptamers, which are single-stranded RNA or DNA sequences, serve as nucleotide equivalents of monoclonal antibodies, displaying high specificity and affinity for target molecules. They are structurally robust, allowing for long-term storage without substantial activity loss. Aptamers find applications in diverse fields such as drug screening, material science, and environmental monitoring. In biomedicine, they are extensively studied for biomarker detection, diagnostics, imaging, and targeted therapy. This comprehensive review focuses on the utility of aptamers in managing infectious diseases, particularly in the realms of diagnostics and therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
12
|
Niedzialkowska E, Runyan LA, Kudryashova E, Egelman EH, Kudryashov DS. Stabilization of F-actin by Salmonella effector SipA resembles the structural effects of inorganic phosphate and phalloidin. Structure 2024; 32:725-738.e8. [PMID: 38518780 PMCID: PMC11162321 DOI: 10.1016/j.str.2024.02.022] [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: 01/02/2024] [Revised: 02/08/2024] [Accepted: 02/26/2024] [Indexed: 03/24/2024]
Abstract
Entry of Salmonella into host enterocytes relies on its pathogenicity island 1 effector SipA. We found that SipA binds to F-actin in a 1:2 stoichiometry with sub-nanomolar affinity. A cryo-EM reconstruction revealed that SipA's globular core binds at the groove between actin strands, whereas the extended C-terminal arm penetrates deeply into the inter-strand space, stabilizing F-actin from within. The unusually strong binding of SipA is achieved by a combination of fast association via the core and very slow dissociation dictated by the arm. Similar to Pi, BeF3, and phalloidin, SipA potently inhibited actin depolymerization by actin depolymerizing factor (ADF)/cofilin, which correlated with increased filament stiffness, supporting the hypothesis that F-actin's mechanical properties contribute to the recognition of its nucleotide state by protein partners. The remarkably strong binding to F-actin maximizes the toxin's effects at the injection site while minimizing global influence on the cytoskeleton and preventing pathogen detection by the host cell.
Collapse
Affiliation(s)
- Ewa Niedzialkowska
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucas A Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA.
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
13
|
Oke MT, D’Costa VM. Functional Divergence of the Paralog Salmonella Effector Proteins SopD and SopD2 and Their Contributions to Infection. Int J Mol Sci 2024; 25:4191. [PMID: 38673776 PMCID: PMC11050076 DOI: 10.3390/ijms25084191] [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: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella enterica is a leading cause of bacterial food-borne illness in humans and is responsible for millions of cases annually. A critical strategy for the survival of this pathogen is the translocation of bacterial virulence factors termed effectors into host cells, which primarily function via protein-protein interactions with host proteins. The Salmonella genome encodes several paralogous effectors believed to have arisen from duplication events throughout the course of evolution. These paralogs can share structural similarities and enzymatic activities but have also demonstrated divergence in host cell targets or interaction partners and contributions to the intracellular lifecycle of Salmonella. The paralog effectors SopD and SopD2 share 63% amino acid sequence similarity and extensive structural homology yet have demonstrated divergence in secretion kinetics, intracellular localization, host targets, and roles in infection. SopD and SopD2 target host Rab GTPases, which represent critical regulators of intracellular trafficking that mediate diverse cellular functions. While SopD and SopD2 both manipulate Rab function, these paralogs display differences in Rab specificity, and the effectors have also evolved multiple mechanisms of action for GTPase manipulation. Here, we highlight this intriguing pair of paralog effectors in the context of host-pathogen interactions and discuss how this research has presented valuable insights into effector evolution.
Collapse
Affiliation(s)
- Mosopefoluwa T. Oke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Vanessa M. D’Costa
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
14
|
Jenniches L, Michaux C, Popella L, Reichardt S, Vogel J, Westermann AJ, Barquist L. Improved RNA stability estimation through Bayesian modeling reveals most Salmonella transcripts have subminute half-lives. Proc Natl Acad Sci U S A 2024; 121:e2308814121. [PMID: 38527194 PMCID: PMC10998600 DOI: 10.1073/pnas.2308814121] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/16/2024] [Indexed: 03/27/2024] Open
Abstract
RNA decay is a crucial mechanism for regulating gene expression in response to environmental stresses. In bacteria, RNA-binding proteins (RBPs) are known to be involved in posttranscriptional regulation, but their global impact on RNA half-lives has not been extensively studied. To shed light on the role of the major RBPs ProQ and CspC/E in maintaining RNA stability, we performed RNA sequencing of Salmonella enterica over a time course following treatment with the transcription initiation inhibitor rifampicin (RIF-seq) in the presence and absence of these RBPs. We developed a hierarchical Bayesian model that corrects for confounding factors in rifampicin RNA stability assays and enables us to identify differentially decaying transcripts transcriptome-wide. Our analysis revealed that the median RNA half-life in Salmonella in early stationary phase is less than 1 min, a third of previous estimates. We found that over half of the 500 most long-lived transcripts are bound by at least one major RBP, suggesting a general role for RBPs in shaping the transcriptome. Integrating differential stability estimates with cross-linking and immunoprecipitation followed by RNA sequencing (CLIP-seq) revealed that approximately 30% of transcripts with ProQ binding sites and more than 40% with CspC/E binding sites in coding or 3' untranslated regions decay differentially in the absence of the respective RBP. Analysis of differentially destabilized transcripts identified a role for ProQ in the oxidative stress response. Our findings provide insights into posttranscriptional regulation by ProQ and CspC/E, and the importance of RBPs in regulating gene expression.
Collapse
Affiliation(s)
- Laura Jenniches
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg97080, Germany
| | - Charlotte Michaux
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg97080, Germany
| | - Linda Popella
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg97080, Germany
| | - Sarah Reichardt
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg97080, Germany
| | - Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg97080, Germany
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg97080, Germany
- Faculty of Medicine, University of Würzburg, Würzburg97080, Germany
| | - Alexander J. Westermann
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg97080, Germany
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg97080, Germany
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg97080, Germany
- Faculty of Medicine, University of Würzburg, Würzburg97080, Germany
- Department of Biology, University of Toronto Mississauga, Mississauga, ONL5L 1C6Canada
| |
Collapse
|
15
|
Nair AV, Singh A, Devasurmutt Y, Rahman SA, Tatu US, Chakravortty D. Spermidine constitutes a key determinant of motility and attachment of Salmonella Typhimurium through a novel regulatory mechanism. Microbiol Res 2024; 281:127605. [PMID: 38232495 DOI: 10.1016/j.micres.2024.127605] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/28/2023] [Accepted: 01/07/2024] [Indexed: 01/19/2024]
Abstract
Spermidine is a poly-cationic molecule belonging to the family of polyamines and is ubiquitously present in all organisms. Salmonella synthesizes, and harbours specialized transporters to import spermidine. A group of polyamines have been shown to assist in Salmonella Typhimurium's virulence and regulation of Salmonella pathogenicity Inslad 1 (SPI-1) genes and stress resistance; however, the mechanism remains elusive. The virulence trait of Salmonella depends on its ability to employ multiple surface structures to attach and adhere to the surface of the target cells before invasion and colonization of the host niche. Our study discovers the mechanism by which spermidine assists in the early stages of Salmonella pathogenesis. For the first time, we report that Salmonella Typhimurium regulates spermidine transport and biosynthesis processes in a mutually inclusive manner. Using a mouse model, we show that spermidine is critical for invasion into the murine Peyer's patches, which further validated our in vitro cell line observation. We show that spermidine controls the mRNA expression of fimbrial (fimA) and non-fimbrial adhesins (siiE, pagN) in Salmonella and thereby assists in attachment to host cell surfaces. Spermidine also regulated the motility through the expression of flagellin genes by enhancing the translation of sigma-28, which features an unusual start codon and a poor Shine-Dalgarno sequence. Besides regulating the formation of the adhesive structures, spermidine tunes the expression of the two-component system BarA/SirA to regulate SPI-1 encoded genes. Thus, our study unravels a novel regulatory mechanism by which spermidine exerts critical functions during Salmonella Typhimurium pathogenesis.
Collapse
Affiliation(s)
- Abhilash Vijay Nair
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Anmol Singh
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Yashas Devasurmutt
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - S A Rahman
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Utpal Shashikant Tatu
- Department of Biochemistry, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India; Adjunct Faculty, School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India.
| |
Collapse
|
16
|
Guo E, Chou SZ, Lara-Tejero M, Galan JE. Cryo-EM structure of the bacterial effector protein SipA bound to F-actin reveals a unique mechanism for filament stabilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572903. [PMID: 38187563 PMCID: PMC10769390 DOI: 10.1101/2023.12.21.572903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The bacterial pathogen Salmonella spp. modulates cellular processes by delivering effector proteins through its type III secretion systems. Among these effectors, SipA facilitates bacterial invasion and promotes intestinal inflammation. The mechanisms by which this effector carries out these functions are incompletely understood although SipA's ability to modulate actin dynamics is central to some of these activities. Here we report the cryo-EM structure of SipA bound to filamentous actin. We show that this effector stabilizes actin filaments through unique interactions of its carboxy terminal domain with four actin subunits. Furthermore, our structure-function studies revealed that SipA's actin-binding activity is independent from its ability to stimulate intestinal inflammation. Overall, these studies illuminate critical aspects of Salmonella pathogenesis, and provide unique insight into the mechanisms by which a bacterial effector modulates actin dynamics.
Collapse
|
17
|
Shuster M, Lyu Z, Augenstreich J, Mathur S, Ganesh A, Ling J, Briken V. Salmonella Typhimurium infection inhibits macrophage IFNβ signaling in a TLR4-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583530. [PMID: 38496427 PMCID: PMC10942315 DOI: 10.1101/2024.03.05.583530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Type I Interferons (IFNs) generally have a protective role during viral infections, but their function during bacterial infections is dependent on the bacterial species. Legionella pneumophila, Shigella sonnei and Mycobacterium tuberculosis can inhibit type I IFN signaling. Here we examined the role of type I IFN, specifically IFNβ, in the context of Salmonella enterica serovar Typhimurium (STm) macrophage infections and the capacity of STm to inhibit type I IFN signaling. We demonstrate that IFNβ has no effect on the intracellular growth of STm in infected bone marrow derived macrophages (BMDMs) derived from C57BL/6 mice. STm infection inhibits IFNβ signaling but not IFNγ signaling in a murine macrophage cell line. We show that this inhibition is independent of the type III and type VI secretion systems expressed by STm and is also independent of bacterial phagocytosis. The inhibition is Toll-like receptor 4 (TLR4)-dependent as the TLR4 ligand, lipopolysaccharide (LPS), alone is sufficient to inhibit IFNβ-mediated signaling and STm-infected, TLR4-deficient BMDMs do not exhibit inhibited IFNβ signaling. In summary, we show that macrophages exposed to STm have reduced IFNβ signaling via crosstalk with TLR4 signaling, and that IFNβ signaling does not affect cell autonomous host defense against STm.
Collapse
Affiliation(s)
- Michael Shuster
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Shrestha Mathur
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Akshaya Ganesh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| |
Collapse
|
18
|
Response to Questions Posed by the Food Safety and Inspection Service: Enhancing Salmonella Control in Poultry Products. J Food Prot 2024; 87:100168. [PMID: 37939849 DOI: 10.1016/j.jfp.2023.100168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 11/10/2023]
|
19
|
Inpanathan S, Ospina-Escobar E, Li VC, Adamji Z, Lackraj T, Cho YH, Porco N, Choy CH, McPhee JB, Botelho RJ. Salmonella actively modulates TFEB in murine macrophages in a growth-phase and time-dependent manner. Microbiol Spectr 2024; 12:e0498122. [PMID: 38051049 PMCID: PMC10783059 DOI: 10.1128/spectrum.04981-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
IMPORTANCE Activation of the host transcription factor TFEB helps mammalian cells adapt to stresses such as starvation and infection by upregulating lysosome, autophagy, and immuno-protective gene expression. Thus, TFEB is generally thought to protect host cells. However, it may also be that pathogenic bacteria like Salmonella orchestrate TFEB in a spatio-temporal manner to harness its functions to grow intracellularly. Indeed, the relationship between Salmonella and TFEB is controversial since some studies showed that Salmonella actively promotes TFEB, while others have observed that Salmonella degrades TFEB and that compounds that promote TFEB restrict bacterial growth. Our work provides a path to resolve these apparent discordant observations since we showed that stationary-grown Salmonella actively delays TFEB after infection, while late-log Salmonella is permissive of TFEB activation. Nevertheless, the exact function of this manipulation remains unclear, but conditions that erase the conditional control of TFEB by Salmonella may be detrimental to the microbe.
Collapse
Affiliation(s)
- Subothan Inpanathan
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Erika Ospina-Escobar
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Vanessa Cruz Li
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Zainab Adamji
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tracy Lackraj
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Youn Hee Cho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Natasha Porco
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Christopher H. Choy
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Joseph B. McPhee
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Roberto J. Botelho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Mohammed BT. Identification and bioinformatic analysis of invA gene of Salmonella in free range chicken. BRAZ J BIOL 2024; 84:e263363. [DOI: 10.1590/1519-6984.263363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Abstract Salmonella is a serious cause of the health issues in human and animal worldwide. Salmonella has been isolated from different biological samples and it considers as the key role in induction of inflammation of gastrointestinal tract which in turn cause diarrhoea in different species. To further understand the involvement of Salmonella in contaminating and infecting fresh eggs and meat of free-range chicken. This study aimed to establish the microbiological and molecular detections of Salmonella in the cloaca of the free-range chicken and to identify predicted biological functions using Kyoto Encyclopedia of Gene and Genomic (KEGG) pathways and protein-protein interaction. Cloacal swabs were collected from free range chicken raised in the local farm in Duhok city. The isolates were cultured and biochemical test performed using XLD and TSI, respectively. Molecular detection and functional annotation of invA gene was carried out using Conventional PCR and bioinformatics approaches. The present study found that Salmonella was detected in 36 out of 86 samples using microbiological methods. To confirm these findings, invA gene was utilised and 9 out of 36 Salmonella isolates have shown a positive signal of invA by agarose gel. In addition, bioinformatic analysis revealed that invA gene was mainly associated with bacterial secretion processes as well as their KEGG terms and Protein-Protein Interaction were involved in bacterial invasion and secretion pathways. These findings suggested that invA gene plays important role in regulating colonization and invasion processes of Salmonella within the gut host in the free range chicken.
Collapse
|
21
|
Wang KC, Jensen PR. Metabolic Analysis of Intracellular Pathogenic Bacteria Using NMR. Methods Mol Biol 2024; 2813:95-105. [PMID: 38888772 DOI: 10.1007/978-1-0716-3890-3_6] [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] [Indexed: 06/20/2024]
Abstract
Pathogen proliferation and virulence depend on available nutrients, and these vary when the pathogen moves from outside of the host cell (extracellular) to the inside of the host cell (intracellular). Nuclear Magnetic Resonance (NMR) is a versatile analytical method, which lends itself for metabolic studies. In this chapter, we describe how 1H NMR can be combined with a cellular infection model to study the metabolic crosstalk between a bacterial pathogen and its host both in the extracellular and intracellular compartments. Central carbon metabolism is highlighted by using glucose labeled with the stable isotope 13C.
Collapse
Affiliation(s)
- Ke-Chuan Wang
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Pernille Rose Jensen
- Center for Hyperpolarization in Magnetic Resonance, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
22
|
Sun X, Yang Y, Meng X, Li J, Liu X, Liu H. PANoptosis: Mechanisms, biology, and role in disease. Immunol Rev 2024; 321:246-262. [PMID: 37823450 DOI: 10.1111/imr.13279] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Cell death can be executed through distinct subroutines. PANoptosis is a unique inflammatory cell death modality involving the interactions between pyroptosis, apoptosis, and necroptosis, which can be mediated by multifaceted PANoptosome complexes assembled via integrating components from other cell death modalities. There is growing interest in the process and function of PANoptosis. Accumulating evidence suggests that PANoptosis occurs under diverse stimuli, for example, viral or bacterial infection, cytokine storm, and cancer. Given the impact of PANoptosis across the disease spectrum, this review briefly describes the relationships between pyroptosis, apoptosis, and necroptosis, highlights the key molecules in PANoptosome formation and PANoptosis activation, and outlines the multifaceted roles of PANoptosis in diseases together with a potential for therapeutic targeting. We also discuss important concepts and pressing issues for future PANoptosis research. Improved understanding of PANoptosis and its mechanisms is crucial for identifying novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Xu Sun
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yanpeng Yang
- Cardiac Care Unit, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiaona Meng
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jia Li
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoli Liu
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Huaimin Liu
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
23
|
Han S, Byun JW, Lee M. Comparative Transcriptomic Analysis of Flagellar-Associated Genes in Salmonella Typhimurium and Its rnc Mutant. J Microbiol 2024; 62:33-48. [PMID: 38182942 DOI: 10.1007/s12275-023-00099-5] [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: 09/27/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a globally recognized foodborne pathogen that affects both animals and humans. Endoribonucleases mediate RNA processing and degradation in the adaptation of bacteria to environmental changes and have been linked to the pathogenicity of S. Typhimurium. Not much is known about the specific regulatory mechanisms of these enzymes in S. Typhimurium, particularly in the context of environmental adaptation. Thus, this study carried out a comparative transcriptomic analysis of wild-type S. Typhimurium SL1344 and its mutant (∆rnc), which lacks the rnc gene encoding RNase III, thereby elucidating the detailed regulatory characteristics that can be attributed to the rnc gene. Global gene expression analysis revealed that the ∆rnc strain exhibited 410 upregulated and 301 downregulated genes (fold-change > 1.5 and p < 0.05), as compared to the wild-type strain. Subsequent bioinformatics analysis indicated that these differentially expressed genes are involved in various physiological functions, in both the wild-type and ∆rnc strains. This study provides evidence for the critical role of RNase III as a general positive regulator of flagellar-associated genes and its involvement in the pathogenicity of S. Typhimurium.
Collapse
Affiliation(s)
- Seungmok Han
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Ji-Won Byun
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Minho Lee
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea.
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
24
|
Abramov VM, Kosarev IV, Machulin AV, Deryusheva EI, Priputnevich TV, Panin AN, Chikileva IO, Abashina TN, Manoyan AM, Akhmetzyanova AA, Blumenkrants DA, Ivanova OE, Papazyan TT, Nikonov IN, Suzina NE, Melnikov VG, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Anti- Salmonella Defence and Intestinal Homeostatic Maintenance In Vitro of a Consortium Containing Limosilactobacillus fermentum 3872 and Ligilactobacillus salivarius 7247 Strains in Human, Porcine, and Chicken Enterocytes. Antibiotics (Basel) 2023; 13:30. [PMID: 38247590 PMCID: PMC10812507 DOI: 10.3390/antibiotics13010030] [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: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Limosilactobacillus fermentum strain 3872 (LF3872) was originally isolated from the breast milk of a healthy woman during lactation and the breastfeeding of a child. Ligilactobacillus salivarius strain 7247 (LS7247) was isolated at the same time from the intestines and reproductive system of a healthy woman. The genomes of these strains contain genes responsible for the production of peptidoglycan-degrading enzymes and factors that increase the permeability of the outer membrane of Gram-negative pathogens. In this work, the anti-Salmonella and intestinal homeostatic features of the LF3872 and LS7247 consortium were studied. A multi-drug resistant (MDR) strain of Salmonella enteritidis (SE) was used in the experiments. The consortium effectively inhibited the adhesion of SE to intact and activated human, porcine, and chicken enterocytes and reduced invasion. The consortium had a bactericidal effect on SE in 6 h of co-culturing. A gene expression analysis of SE showed that the cell-free supernatant (CFS) of the consortium inhibited the expression of virulence genes critical for the colonization of human and animal enterocytes. The CFS stimulated the production of an intestinal homeostatic factor-intestinal alkaline phosphatase (IAP)-in Caco-2 and HT-29 enterocytes. The consortium decreased the production of pro-inflammatory cytokines IL-8, TNF-α, and IL-1β, and TLR4 mRNA expression in human and animal enterocytes. It stimulated the expression of TLR9 in human and porcine enterocytes and stimulated the expression of TLR21 in chicken enterocytes. The consortium also protected the intestinal barrier functions through the increase of transepithelial electrical resistance (TEER) and the inhibition of paracellular permeability in the monolayers of human and animal enterocytes. The results obtained suggest that a LF3872 and LS7247 consortium can be used as an innovative feed additive to reduce the spread of MDR SE among the population and farm animals.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Irina O. Chikileva
- Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ashot M. Manoyan
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Anna A. Akhmetzyanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Dmitriy A. Blumenkrants
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Olga E. Ivanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | | | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia;
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia; (V.S.K.); (V.K.S.)
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
25
|
Niedzialkowska E, Runyan LA, Kudryashova E, Egelman EH, Kudryashov DS. Stabilization of F-actin by Salmonella effector SipA resembles the structural effects of inorganic phosphate and phalloidin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.26.573373. [PMID: 38234808 PMCID: PMC10793455 DOI: 10.1101/2023.12.26.573373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Entry of Salmonella into host enterocytes strictly relies on its pathogenicity island 1 effector SipA. We found that SipA binds to F-actin in a unique mode in a 1:2 stoichiometry with picomolar affinity. A cryo-EM reconstruction revealed that SipA's globular core binds at the grove between actin strands, whereas the extended C-terminal arm penetrates deeply into the inter-strand space, stabilizing F-actin from within. The unusually strong binding of SipA is achieved via a combination of fast association via the core and very slow dissociation dictated by the arm. Similarly to Pi, BeF3, and phalloidin, SipA potently inhibited actin depolymerization by ADF/cofilin, which correlated with the increased filament stiffness, supporting the hypothesis that F-actin's mechanical properties contribute to the recognition of its nucleotide state by protein partners. The remarkably strong binding to F-actin maximizes the toxin's effects at the injection site while minimizing global influence on the cytoskeleton and preventing pathogen detection by the host cell.
Collapse
Affiliation(s)
- Ewa Niedzialkowska
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucas A. Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
26
|
Berni M, Bolzoni L, Menozzi I, Dodi A, Bracchi C, Morganti M, Scaltriti E, Pongolini S, Tambassi M. Salmonella Derby adaptation to swine and simultaneous attenuation for humans go through decay of Salmonella Pathogenicity Island I. Microbiol Spectr 2023; 11:e0189923. [PMID: 37800927 PMCID: PMC10715017 DOI: 10.1128/spectrum.01899-23] [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: 05/19/2023] [Accepted: 08/09/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE This study integrated population data with in vitro assessment of virulence phenotypes to unveil that a considerable part of the global population of Salmonella Derby is evolving to enhance its host adaptation to the swine host and that this evolution is simultaneously increasing its attenuation for humans. The study shows that the fixation of deleterious mutations in SPI-1 has a role in this process. This evidence indicates that SPI-1 has a key role for S. Derby virulence in humans but not for its circulation in swine. The results show that genes generally considered essential for Salmonella pathogenesis do not play the same key role for all Salmonella serovars or lineages and/or all hosts. The study helps in understanding the molecular mechanisms underlying the ecology and host adaptation of Salmonella showing that the adaptation process can vary for different types of Salmonella and hosts.
Collapse
Affiliation(s)
- Melissa Berni
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Luca Bolzoni
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Ilaria Menozzi
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Alessandra Dodi
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Chiara Bracchi
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Marina Morganti
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Erika Scaltriti
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Stefano Pongolini
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| | - Martina Tambassi
- Risk Analysis and Genomic Epidemiology Unit, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna (IZSLER), Parma, Italy
| |
Collapse
|
27
|
Pradhan J, Pradhan D, Sahu JK, Mishra S, Mallick S, Das S, Negi VD. A novel rspA gene regulates biofilm formation and virulence of Salmonella Typhimurium. Microb Pathog 2023; 185:106432. [PMID: 37926364 DOI: 10.1016/j.micpath.2023.106432] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Salmonella spp. are facultative anaerobic, Gram-negative, rod-shaped bacteria and belongs to the Enterobacteriaceae family. Although much has been known about Salmonella pathogenesis, the functional characterizations of certain genes are yet to be explored. The rspA (STM14_1818) is one such gene with putative dehydratase function, and its role in pathogenesis is unknown. The background information showed that rspA gene is upregulated in Salmonella when it resides inside macrophages, which led us to investigate its role in Salmonella pathogenesis. We generated the rspA knockout strain and complement strain in S. Typhimurium 14028. Ex-vivo and in-vivo infectivity was looked at macrophage and epithelial cell lines and Caenorhabditis elegans (C. elegans). The mutant strain differentially formed the biofilm at different temperatures by altering the expression of genes involved in the synthesis of cellulose and curli. Besides, the mutant strain is hyperproliferative intracellularly and showed increased bacterial burden in C. elegans. The mutant strain became more infectious and lethal, causing faster death of the worms than the wild type, and also modulates the worm's innate immunity. Thus, we found that the rspA deletion mutant was more pathogenic. In this study, we concluded that the rspA gene differentially regulates the biofilm formation in a temperature dependent manner by modulating the genes involved in the synthesis of cellulose and curli and negatively regulates the Salmonella virulence for longer persistence inside the host.
Collapse
Affiliation(s)
- Jasmin Pradhan
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Diana Pradhan
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Jugal Kishor Sahu
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Satyajit Mishra
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Swarupa Mallick
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Surajit Das
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Vidya Devi Negi
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
28
|
Sutton K, Nash T, Sives S, Borowska D, Mitchell J, Vohra P, Stevens MP, Vervelde L. Disentangling the innate immune responses of intestinal epithelial cells and lamina propria cells to Salmonella Typhimurium infection in chickens. Front Microbiol 2023; 14:1258796. [PMID: 37854334 PMCID: PMC10579587 DOI: 10.3389/fmicb.2023.1258796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (STm) is a major foodborne pathogen and poultry are a key reservoir of human infections. To understand the host responses to early stages of Salmonella infection in poultry, we infected 2D and 3D enteroids, the latter of which contains leukocytes, neurons, and mesenchymal cells that are characteristic of the lamina propria. We infected these enteroids with wild-type (WT STm), a non-invasive mutant lacking the prgH gene (ΔprgH STm), or treated them with STm lipopolysaccharide (LPS) and analyzed the expression of innate immune related genes by qPCR at 4 and 8 h. The localization of the tight junction protein, ZO-1, expression was disrupted in WT STm infected enteroids but not ΔprgH STm or LPS treated enteroids, suggesting a loss of epithelial barrier integrity. The innate immune response to LPS was more pronounced in 2D enteroids compared to 3D enteroids and by 8 hpi, the response in 3D enteroids was almost negligible. However, when STm adhered to or invaded the enteroids, both 2D and 3D enteroids exhibited an upregulation of inflammatory responses. The presence of lamina propria cells in 3D enteroids resulted in the unique expression of genes associated with immune functions involved in regulating inflammation. Moreover, 2D and 3D enteroids showed temporal differences in response to bacterial invasion or adherence. At 8 hpi, innate responses in 3D but not 2D enteroids continued to increase after infection with WT STm, whereas the responses to the non-invasive strain decreased at 8 hpi in both 2D and 3D enteroids. In conclusion, STm infection of chicken enteroids recapitulated several observations from in vivo studies of Salmonella-infected chickens, including altered epithelial barrier integrity based on ZO-1 expression and inflammatory responses. Our findings provide evidence that Salmonella-infected enteroids serve as effective models for investigating host-pathogen interactions and exploring the molecular mechanisms of microbial virulence although the 3D model mimics the host more accurately due to the presence of a lamina propria.
Collapse
Affiliation(s)
- Kate Sutton
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Tessa Nash
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Samantha Sives
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Dominika Borowska
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Jordan Mitchell
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Prerna Vohra
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark P. Stevens
- Division of Bacteriology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Lonneke Vervelde
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
29
|
López-Yglesias AH, Lu CC, Lai MA, Quarles EK, Zhao X, Hajjar AM, Smith KD. FlgM is required to evade NLRC4-mediated host protection against flagellated Salmonella. Infect Immun 2023; 91:e0025523. [PMID: 37638725 PMCID: PMC10501211 DOI: 10.1128/iai.00255-23] [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: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/29/2023] Open
Abstract
Salmonella enterica serovar Typhimurium is a leading cause of gastroenteritis worldwide and a deadly pathogen in children, immunocompromised patients, and the elderly. Salmonella induces innate immune responses through the NLRC4 inflammasome, which has been demonstrated to have distinct roles during systemic and mucosal detections of flagellin and non-flagellin molecules. We hypothesized that NLRC4 recognition of Salmonella flagellin is the dominant protective pathway during infection. To test this hypothesis, we used wild-type, flagellin-deficient, and flagellin-overproducing Salmonella to establish the role of flagellin in mediating NLRC4-dependent host resistance during systemic and mucosal infections in mice. We observed that during the systemic phase of infection, Salmonella efficiently evades NLRC4-mediated innate immunity. During mucosal Salmonella infection, flagellin recognition by the NLRC4 inflammasome pathway is the dominant mediator of protective innate immunity. Deletion of flgM results in constitutive expression of flagellin and severely limits systemic and mucosal Salmonella infections in an NLRC4 inflammasome-dependent manner. These data establish that recognition of Salmonella's flagellin by the NLRC4 inflammasome during mucosal infection is the dominant innate protective pathway for host resistance against the enteric pathogen and that FlgM-mediated evasion of the NLRC4 inflammasome enhances virulence and intestinal tissue destruction.
Collapse
Affiliation(s)
| | - Chun-Chi Lu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Marvin A. Lai
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ellen K. Quarles
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Xiaodan Zhao
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington, USA
| | - Kelly D. Smith
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
30
|
Chatterjee R, Chaudhuri D, Setty SRG, Chakravortty D. Deceiving the big eaters: Salmonella Typhimurium SopB subverts host cell xenophagy in macrophages via dual mechanisms. Microbes Infect 2023; 25:105128. [PMID: 37019426 DOI: 10.1016/j.micinf.2023.105128] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Salmonella, a stealthy facultative intracellular pathogen, utilises an array of host immune evasion strategies. This facilitates successful survival via replicative niche establishment in otherwise hostile environments such as macrophages. Salmonella survives in and utilises macrophages for effective dissemination, ultimately leading to systemic infection. Bacterial xenophagy or macro-autophagy is an important host defense mechanism in macrophages. Here, we report for the first time that the Salmonella pathogenicity island-1 (SPI-1) effector SopB is involved in subverting host autophagy via dual mechanisms. SopB is a phosphoinositide phosphatase capable of altering the phosphoinositide dynamics of the host cell. Here, we demonstrate that SopB mediates escape from autophagy by inhibiting the terminal fusion of Salmonella-containing vacuoles (SCVs) with lysosomes and/or autophagosomes. We also report that SopB downregulates overall lysosomal biogenesis by modulating the Akt-transcription factor EB (TFEB) axis via restricting the latter's nuclear localisation. TFEB is a master regulator of lysosomal biogenesis and autophagy. This reduces the overall lysosome content inside host macrophages, further facilitating the survival of Salmonella in macrophages and systemic dissemination of Salmonella.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Debalina Chaudhuri
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India; Indian Institute of Science Research and Education, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
31
|
Pillay TD, Hettiarachchi SU, Gan J, Diaz-Del-Olmo I, Yu XJ, Muench JH, Thurston TL, Pearson JS. Speaking the host language: how Salmonella effector proteins manipulate the host. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001342. [PMID: 37279149 PMCID: PMC10333799 DOI: 10.1099/mic.0.001342] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Salmonella injects over 40 virulence factors, termed effectors, into host cells to subvert diverse host cellular processes. Of these 40 Salmonella effectors, at least 25 have been described as mediating eukaryotic-like, biochemical post-translational modifications (PTMs) of host proteins, altering the outcome of infection. The downstream changes mediated by an effector's enzymatic activity range from highly specific to multifunctional, and altogether their combined action impacts the function of an impressive array of host cellular processes, including signal transduction, membrane trafficking, and both innate and adaptive immune responses. Salmonella and related Gram-negative pathogens have been a rich resource for the discovery of unique enzymatic activities, expanding our understanding of host signalling networks, bacterial pathogenesis as well as basic biochemistry. In this review, we provide an up-to-date assessment of host manipulation mediated by the Salmonella type III secretion system injectosome, exploring the cellular effects of diverse effector activities with a particular focus on PTMs and the implications for infection outcomes. We also highlight activities and functions of numerous effectors that remain poorly characterized.
Collapse
Affiliation(s)
- Timesh D. Pillay
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Sahampath U. Hettiarachchi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Jiyao Gan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Ines Diaz-Del-Olmo
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
| | - Xiu-Jun Yu
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
| | - Janina H. Muench
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Teresa L.M. Thurston
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Jaclyn S. Pearson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
32
|
Thomas C, Methner U, Marz M, Linde J. Oxford nanopore technologies-a valuable tool to generate whole-genome sequencing data for in silico serotyping and the detection of genetic markers in Salmonella. Front Vet Sci 2023; 10:1178922. [PMID: 37323838 PMCID: PMC10267320 DOI: 10.3389/fvets.2023.1178922] [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: 03/03/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023] Open
Abstract
Bacteria of the genus Salmonella pose a major risk to livestock, the food economy, and public health. Salmonella infections are one of the leading causes of food poisoning. The identification of serovars of Salmonella achieved by their diverse surface antigens is essential to gain information on their epidemiological context. Traditionally, slide agglutination has been used for serotyping. In recent years, whole-genome sequencing (WGS) followed by in silico serotyping has been established as an alternative method for serotyping and the detection of genetic markers for Salmonella. Until now, WGS data generated with Illumina sequencing are used to validate in silico serotyping methods. Oxford Nanopore Technologies (ONT) opens the possibility to sequence ultra-long reads and has frequently been used for bacterial sequencing. In this study, ONT sequencing data of 28 Salmonella strains of different serovars with epidemiological relevance in humans, food, and animals were taken to investigate the performance of the in silico serotyping tools SISTR and SeqSero2 compared to traditional slide agglutination tests. Moreover, the detection of genetic markers for resistance against antimicrobial agents, virulence, and plasmids was studied by comparing WGS data based on ONT with WGS data based on Illumina. Based on the ONT data from flow cell version R9.4.1, in silico serotyping achieved an accuracy of 96.4 and 92% for the tools SISTR and SeqSero2, respectively. Highly similar sets of genetic markers comparing both sequencing technologies were identified. Taking the ongoing improvement of basecalling and flow cells into account, ONT data can be used for Salmonella in silico serotyping and genetic marker detection.
Collapse
Affiliation(s)
- Christine Thomas
- Institute of Bacterial Infections and Zoonoses, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute, Jena, Germany
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
| | - Ulrich Methner
- Institute of Bacterial Infections and Zoonoses, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute, Jena, Germany
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
| | - Jörg Linde
- Institute of Bacterial Infections and Zoonoses, Federal Research Institute for Animal Health, Friedrich-Loeffler-Institute, Jena, Germany
| |
Collapse
|
33
|
Sarma A, Gunasekaran D, Phukan H, Baby A, Hariharan S, De AK, Bhattacharya D, Natesan S, Tennyson J, Madanan MG. Leptospiral imelysin (LIC_10713) is secretory, immunogenic and binds to laminin, fibronectin, and collagen IV. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12573-6. [PMID: 37227474 DOI: 10.1007/s00253-023-12573-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/26/2023]
Abstract
Leptospirosis is a widespread zoonotic disease caused by pathogenic Leptospira. Early and accurate diagnosis is the prime step in managing the disease. Secretory proteins of Leptospira remain distinguished for diagnosis due to their availability as soluble proteins in the serum and their interaction with the host immune response due to their extracellular presence. This study presents the cloning, expression, purification, and characterization of imelysin or LruB (LIC_10713), a putative leptospiral protein. We report that the localization of imelysin showed its presence in the inner membrane and in the culture supernatant. The imelysin was upregulated under in vitro physiological conditions of infection. The LIC_10713 interacted significantly with laminin, fibronectin, collagen type I, and collagen type IV in a dose-dependent manner. Phylogenetic analysis showed that LIC_10713 is predominately found in the pathogenic species of Leptospira, and the GxHxxE motif of imelysin-like proteins is represented as the amino acid sequence GWHAIE. Also, immunoglobulins in leptospirosis-infected patients recognize recombinant-LIC_10713 with 100% specificity and 90.9% sensitivity. The secretion nature, abundance, upregulation, binding to ECM components, and immunogenicity determine LIC_10713 as an important molecule that can be used as an anti-leptospirosis measure. KEY POINTS: • The imelysin-like protein (LIC_10713) of Leptospira is a secretory protein • The protein LIC_10713 can bind ECM molecules • The LIC_10713 is mainly found in pathogenic leptospires • The anti-LIC_10713 antibody from human serum can detect the r-LIC_10713.
Collapse
Affiliation(s)
- Abhijit Sarma
- Department of Biochemistry, ICMR - Regional Medical Research Centre, Port Blair, 744103, Andaman and Nicobar Islands, India
| | - Dhandapani Gunasekaran
- Department of Biochemistry, ICMR - Regional Medical Research Centre, Port Blair, 744103, Andaman and Nicobar Islands, India
| | - Homen Phukan
- Department of Biochemistry, ICMR - Regional Medical Research Centre, Port Blair, 744103, Andaman and Nicobar Islands, India
| | - Akhil Baby
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamraj University, Madurai, Tamil Nadu, 625021, India
| | - Suneetha Hariharan
- Department of Biochemistry, ICMR - Regional Medical Research Centre, Port Blair, 744103, Andaman and Nicobar Islands, India
| | - Arun Kumar De
- Division of Animal Science, ICAR- Central Island Agricultural Research Institute, Port Blair, 744105, Andaman and Nicobar Islands, India
| | - Debasis Bhattacharya
- Division of Animal Science, ICAR- Central Island Agricultural Research Institute, Port Blair, 744105, Andaman and Nicobar Islands, India
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamraj University, Madurai, Tamil Nadu, 625021, India
| | - Jebasingh Tennyson
- Department of Plant Sciences, School of Biological Sciences, Madurai Kamraj University, Madurai, Tamil Nadu, 625021, India
| | | |
Collapse
|
34
|
Davidson A, Hume PJ, Greene NP, Koronakis V. Salmonella invasion of a cell is self-limiting due to effector-driven activation of N-WASP. iScience 2023; 26:106643. [PMID: 37168569 PMCID: PMC10164908 DOI: 10.1016/j.isci.2023.106643] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 04/06/2023] [Indexed: 05/13/2023] Open
Abstract
Salmonella Typhimurium drives uptake into non-phagocytic host cells by injecting effector proteins that reorganize the actin cytoskeleton. The host actin regulator N-WASP has been implicated in bacterial entry, but its precise role is not clear. We demonstrate that Cdc42-dependent N-WASP activation, instigated by the Cdc42-activating effector SopE2, strongly impedes Salmonella uptake into host cells. This inhibitory pathway is predominant later in invasion, with the ubiquitin ligase activity of the effector SopA specifically interfering with negative Cdc42-N-WASP signaling at early stages. The cell therefore transitions from being susceptible to invasion, into a state almost completely recalcitrant to bacterial uptake, providing a mechanism to limit the number of internalized Salmonella. Our work raises the possibility that Cdc42-N-WASP, known to be activated by numerous bacterial and viral species during infection and commonly assumed to promote pathogen uptake, is used to limit the entry of multiple pathogens.
Collapse
Affiliation(s)
| | - Peter J. Hume
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Vassilis Koronakis
- Department of Pathology, University of Cambridge, Cambridge, UK
- Corresponding author
| |
Collapse
|
35
|
Kumar Panda R, Darshana Patra S, Kumar Mohakud N, Ranjan Sahu B, Ghosh M, Misra N, Suar M. Draft genome of clinical isolate Salmonella enterica Typhimurium ms204 from Odisha, India, reveals multi drug resistance and decreased virulent gene expression. Gene 2023; 863:147248. [PMID: 36738898 DOI: 10.1016/j.gene.2023.147248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Salmonellosis, a food-borne illnesses caused by enteropathogenic bacterium Salmonella spp., is a continuous concern in both developed and developing countries. This study was carried out to perform an in-depth examination of an MDR Salmonella strain isolated from gastroenteritis patients in Odisha, India, in order to understand the genomic architecture, distribution of pathogenic island regions, and virulence factor diversity. Fecal samples were obtained from individuals with acute gastroenteritis and further subjected to panel of biochemical tests. The IlluminaHiSeq X sequencer system was used to generate whole-genome sequencing. The draft genome was submitted to gene prediction and annotation using RAST annotation system. Pathogenicity Island database and bioinformatics pipeline were used to find Salmonella pathogenicity islands (SPI) from the built scaffold. The gene expression in SPI1 and SPI2 encoded regions was investigated using qRT-PCR. The taxonomic position of Salmonella enterica subsp. enterica serovar Typhimurium was validated by serotype analysis and 16S rRNA based phylogenetic analysis. The de-novo genome assembly showed total length of 5,034,110 bp and produced 37 contigs. There are nine prophage areas, comprising of 12 regions and scaffold 8 contained a single plasmid, IncFIB. The isolate contains six known SPI genes content which was shown to be largely conserved from SPI1 to SPI2. We identified the sit ABCD cluster regulatory cascade and acquired antibiotic resistance genes in S. enterica Typhimurium ms204. Further research may aid in the correct diagnosis and monitoring of MDR Salmonella strains with a variety of physiological activities.
Collapse
Affiliation(s)
| | | | - Nirmal Kumar Mohakud
- Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar 751024, India
| | - Bikash Ranjan Sahu
- Department of Zoology, Centurion University of Technology and Management, India
| | - Mrinmoy Ghosh
- KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar 751024, India
| | - Namrata Misra
- KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar 751024, India
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, India; KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar 751024, India.
| |
Collapse
|
36
|
Shin H, La TM, Lee HJ, Kim T, Song SU, Park GH, Choi IS, Park SY, Lee JB, Lee SW. Characteristics of a Temperature-Sensitive Mutant Strain of Salmonella Enteritidis and Its Potential as a Live Vaccine Candidate. Vet Sci 2023; 10:vetsci10050313. [PMID: 37235396 DOI: 10.3390/vetsci10050313] [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: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Salmonella Enteritidis is a common foodborne pathogen transmitted through poultry products, which are its main carriers. Poultry are vaccinated against Salmonella Enteritidis in many countries, despite the absence of clinical symptoms, using commercially available live-attenuated vaccines. We previously constructed a highly attenuated temperature-sensitive (ts) Salmonella Enteritidis mutant, 2S-G10. In the present study, we describe the construction and attenuation-associated characteristics of 2S-G10. We infected 1-day-old chicks with 2S-G10 and the parental strains to evaluate the attenuation. One week after infection, 2S-G10 was not detected in the liver, cecum, or cecal tonsil tissues of the orally inoculated chicks, contrary to the parental strain. This indicates that 2S-G10 was highly attenuated when compared to the parental stain. In vitro experiments revealed the inability of 2S-G10 to grow at the normal body temperature of chickens and invade chicken liver epithelial cells. Moreover, single nucleotide polymorphism (SNP) analysis between the complete genome sequence of 2S-G10 and its parental strain revealed SNPs in bcsE, recG, rfaF, and pepD_1 genes, which are involved in epithelial cell invasion and persistence in host systems, growth, lipopolysaccharide core biosynthesis, and cellular survival under heat stress, respectively. These potential characteristics are consistent with the findings of in vitro experiments. Conclusively, chemical treatment-induced random genetic mutations highly attenuated 2S-G10, implying its potential to be developed as a novel live-attenuated vaccine against Salmonella Enteritidis.
Collapse
Affiliation(s)
- Hyunjin Shin
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Tae-Min La
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hong-Jae Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Taesoo Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Seung-Un Song
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gyu-Hyung Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - In-Soo Choi
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Seung-Yong Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Joong-Bok Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Sang-Won Lee
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
37
|
Panwar S, Kumari S, Verma J, Bakshi S, Narendrakumar L, Paul D, Das B. Toxin-linked mobile genetic elements in major enteric bacterial pathogens. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e5. [PMID: 39295911 PMCID: PMC11406385 DOI: 10.1017/gmb.2023.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 12/31/2022] [Accepted: 02/24/2023] [Indexed: 09/21/2024]
Abstract
One of the fascinating outcomes of human microbiome studies adopting multi-omics technology is its ability to decipher millions of microbial encoded functions in the most complex and crowded microbial ecosystem, including the human gastrointestinal (GI) tract without cultivating the microbes. It is well established that several functions that modulate the human metabolism, nutrient assimilation, immunity, infections, disease severity and therapeutic efficacy of drugs are mostly of microbial origins. In addition, these microbial functions are dynamic and can disseminate between microbial taxa residing in the same ecosystem or other microbial ecosystems through horizontal gene transfer. For clinicians and researchers alike, understanding the toxins, virulence factors and drug resistance traits encoded by the microbes associated with the human body is of utmost importance. Nevertheless, when such traits are genetically linked with mobile genetic elements (MGEs) that make them transmissible, it creates an additional burden to public health. This review mainly focuses on the functions of gut commensals and the dynamics and crosstalk between commensal and pathogenic bacteria in the gut. Also, the review summarises the plethora of MGEs linked with virulence genes present in the genomes of various enteric bacterial pathogens, which are transmissible among other pathogens and commensals.
Collapse
Affiliation(s)
- Shruti Panwar
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Shashi Kumari
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyoti Verma
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Susmita Bakshi
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Lekshmi Narendrakumar
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Deepjyoti Paul
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
38
|
Serotype Occurrence, Virulence Profiles, Antimicrobial Resistance and Molecular Characterization of Salmonella Isolated from Hospitalized Patients with Gastroenteritis in Great Tunisia between 2010 and 2020. Antibiotics (Basel) 2023; 12:antibiotics12030526. [PMID: 36978394 PMCID: PMC10044041 DOI: 10.3390/antibiotics12030526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Non-typhoid Salmonella is one of the major causes of food-borne infections worldwide. The aim of the current study is to determine the serotype occurrence, virulence factors and antimicrobial resistance patterns of Salmonella isolated from hospitalized patients. The identification of Salmonella strains was performed according to REMIC, 2018. The susceptibility of Salmonella isolates was assessed against 20 antimicrobials using the disk diffusion method. Some virulence and antimicrobial resistance genes were identified using PCR. Among the 61 isolated Salmonella strains, seven serotypes were identified and all were positive for the virulence genes invA, mgtC and sirA. Critical resistance rates (>40%) were detected for tetracycline, nalidixic acid, amoxicillin and fluoroquinolones. However, resistances to ertapenem, ceftazidim, aztreonam and colistin were null. In addition, 33% of the isolated strains were multidrug-resistant (MDR). Moreover, 80% and 60% of S. Kentucky isolates were identified as fluoroquinolone-resistant and MDR strains, respectively. The qnrB gene was amplified in 63.2% of fluoroquinolone-resistant strains. The dfrA1 gene was identified in 20% (4/20) of the trimethoprim-sulfamethoxazole resistant strains and the integrase Class 2 gene was amplified in only 8.2% (5/61) of the isolates. Our findings highlight the emergence of MDR Salmonella isolates. A rationalization of antimicrobial use is urgently recommended in both human and veterinary medicine.
Collapse
|
39
|
SARIÇAM İNCE S, AKAN M. Molecular characterization of virulence genes in poultry-originated Salmonella Enteritidis and Salmonella Typhimurium. ANKARA ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2023. [DOI: 10.33988/auvfd.1157022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Salmonella Enteritidis and Salmonella Typhimurium are the most common serovars observed in human salmonellosis while contaminated poultry products are the major source of Salmonella transmission to humans. Therefore, high pathogenicity of poultry-originated S. Enteritidis and S. Typhimurium strains poses a serious risk to human health. This study analyzed the virulence genes of broiler chicken-originated S. Enteritidis and S. Typhimurium strains. SipA, sipD, sopB, sopD, sopE, sopE2, sitC, sifA, ssaR, spvC and pefA genes were investigated in a total of 137 strains consisting of 105 S. Enteritidis and 32 S. Typhimurium. Nine strains (6.6%) had all genes. No negative strain was detected for all genes. SopE was found in all strains (100%). SitC (89.1%), ssaR (83.9%), sipA (70.1%), sipD (73.0%), sopE2 (68.6%), spvC (68.6%) and pefA (73.0%) were also highly prevalent. Noticeable differences were observed between serovars in terms of sopE2, spvC and pefA prevalence: 77.1%, 80% and 82.9%, respectively, of S. Enteritidis strains were sopE2, spvC and pefA positive while 40.6%, 31.3% and 40.6% of S. Typhimurium strains were positive. This finding indicates that S. Enteritidis is more frequent than S. Typhimurium in poultry populations thanks to higher virulence. Based on virulence gene distribution, the strains were divided into 44 different virulence genotypes, with the major genotype 4 (15.3%) carrying 8 of the 11 genes. The majority of strains (75.9%) were positive for at least 6 genes. S. Enteritidis and S. Typhimurium strains were highly virulent and pose a threat as a zoonotic infection.
Collapse
Affiliation(s)
| | - Mehmet AKAN
- ANKARA UNIVERSITY, ANKARA FACULTY OF VETERINARY MEDICINE
| |
Collapse
|
40
|
Yuan H, Zhou L, Chen Y, You J, Hu H, Li Y, Huang R, Wu S. Salmonella effector SopF regulates PANoptosis of intestinal epithelial cells to aggravate systemic infection. Gut Microbes 2023; 15:2180315. [PMID: 36803521 PMCID: PMC9980482 DOI: 10.1080/19490976.2023.2180315] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
SopF, a newly discovered effector secreted by Salmonella pathogenicity island-1 type III secretion system (T3SS1), was reported to target phosphoinositide on host cell membrane and aggravate systemic infection, while its functional relevance and underlying mechanisms have yet to be elucidated. PANoptosis (pyroptosis, apoptosis, and necroptosis) of intestinal epithelial cells (IECs) has been characterized as a pivotal host defense to limit the dissemination of foodborne pathogens, whereas the effect of SopF on IECs PANoptosis induced by Salmonella is rather limited. Here, we show that SopF can attenuate intestinal inflammation and suppress IECs expulsion to promote bacterial dissemination in mice infected with Salmonella enterica serovar Typhimurium (S. Typhimurium). We revealed that SopF could activate phosphoinositide-dependent protein kinase-1 (PDK1) to phosphorylate p90 ribosomal S6 kinase (RSK) which down-regulated Caspase-8 activation. Caspase-8 inactivated by SopF resulted in inhibition of pyroptosis and apoptosis, but promotion of necroptosis. The administration of both AR-12 (PDK1 inhibitor) and BI-D1870 (RSK inhibitor) potentially overcame Caspase-8 blockade and subverted PANoptosis challenged by SopF. Collectively, these findings demonstrate that this virulence strategy elicited by SopF aggregates systemic infection via modulating IEC PANoptosis through PDK1-RSK signaling, which throws light on novel functions of bacterial effectors, as well as a mechanism employed by pathogens to counteract host immune defense.
Collapse
Affiliation(s)
- Haibo Yuan
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China,Department of Medical Technology, Suzhou Vocational Health College, Suzhou, China
| | - Liting Zhou
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China,Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine
| | - Yilin Chen
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiayi You
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hongye Hu
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yuanyuan Li
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China,Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine
| | - Rui Huang
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China,Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine
| | - Shuyan Wu
- Department of Medical Microbiology, School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China,Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine,CONTACT Shuyan Wu; Rui Huang ; Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, No. 199, Ren Ai Road, Suzhou, Jiangsu215123, PR China
| |
Collapse
|
41
|
Transcriptional insight into the effect of benzalkonium chloride on resistance and virulence potential in Salmonella Typhimurium. Microbiol Res 2023; 266:127240. [DOI: 10.1016/j.micres.2022.127240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/26/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022]
|
42
|
Romero-Calle DX, Pedrosa-Silva F, Tomé LMR, Sousa TJ, de Oliveira Santos LTS, de Carvalho Azevedo VA, Brenig B, Benevides RG, Venancio TM, Billington C, Góes-Neto A. Hybrid Genomic Analysis of Salmonella enterica Serovar Enteritidis SE3 Isolated from Polluted Soil in Brazil. Microorganisms 2022; 11:111. [PMID: 36677403 PMCID: PMC9861973 DOI: 10.3390/microorganisms11010111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
In Brazil, Salmonella enterica serovar Enteritidis is a significant health threat. Salmonella enterica serovar Enteritidis SE3 was isolated from soil at the Subaé River in Santo Amaro, Brazil, a region contaminated with heavy metals and organic waste. Illumina HiSeq and Oxford Nanopore Technologies MinION sequencing were used for de novo hybrid assembly of the Salmonella SE3 genome. This approach yielded 10 contigs with 99.98% identity with S. enterica serovar Enteritidis OLF-SE2-98984-6. Twelve Salmonella pathogenic islands, multiple virulence genes, multiple antimicrobial gene resistance genes, seven phage defense systems, seven prophages and a heavy metal resistance gene were encoded in the genome. Pangenome analysis of the S. enterica clade, including Salmonella SE3, revealed an open pangenome, with a core genome of 2137 genes. Our study showed the effectiveness of a hybrid sequence assembly approach for environmental Salmonella genome analysis using HiSeq and MinION data. This approach enabled the identification of key resistance and virulence genes, and these data are important to inform the control of Salmonella and heavy metal pollution in the Santo Amaro region of Brazil.
Collapse
Affiliation(s)
- Danitza Xiomara Romero-Calle
- Postgraduate Program in Biotechnology, State University of Feira de Santana (UEFS), Av. Transnordestina S/N, Feira de Santana 44036-900, BA, Brazil
- Molecular and Computational Biology of Fungi Laboratory, Department of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Department of Biological Sciences, Feira de Santana State University (UEFS), Feira de Santana 44036-900, BA, Brazil
| | - Francisnei Pedrosa-Silva
- Laboratory of Chemistry, Function of Proteins and Peptides, Center for Biosciences and Biotechnology, Darcy Ribeiro North Fluminense State University (UENF), Campos dos Goytacazes 28013-602, RJ, Brazil
| | - Luiz Marcelo Ribeiro Tomé
- Molecular and Computational Biology of Fungi Laboratory, Department of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Thiago J. Sousa
- Laboratory of Cellular and Molecular Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | | | - Vasco Ariston de Carvalho Azevedo
- Laboratory of Cellular and Molecular Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Bertram Brenig
- Institute of Veterinary Medicine, Burckhardtweg, University of Göttingen, 37073 Göttingen, Germany
| | - Raquel Guimarães Benevides
- Postgraduate Program in Biotechnology, State University of Feira de Santana (UEFS), Av. Transnordestina S/N, Feira de Santana 44036-900, BA, Brazil
- Department of Biological Sciences, Feira de Santana State University (UEFS), Feira de Santana 44036-900, BA, Brazil
| | - Thiago M. Venancio
- Laboratory of Chemistry, Function of Proteins and Peptides, Center for Biosciences and Biotechnology, Darcy Ribeiro North Fluminense State University (UENF), Campos dos Goytacazes 28013-602, RJ, Brazil
| | - Craig Billington
- Health & Environment Group, Institute of Environmental Sciences and Research, P.O. Box 29-181, Christchurch 8540, New Zealand
| | - Aristóteles Góes-Neto
- Postgraduate Program in Biotechnology, State University of Feira de Santana (UEFS), Av. Transnordestina S/N, Feira de Santana 44036-900, BA, Brazil
- Molecular and Computational Biology of Fungi Laboratory, Department of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Department of Biological Sciences, Feira de Santana State University (UEFS), Feira de Santana 44036-900, BA, Brazil
- Laboratory of Cellular and Molecular Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
43
|
Cohn AR, Orsi RH, Carroll LM, Liao J, Wiedmann M, Cheng RA. Salmonella enterica serovar Cerro displays a phylogenetic structure and genomic features consistent with virulence attenuation and adaptation to cattle. Front Microbiol 2022; 13:1005215. [PMID: 36532462 PMCID: PMC9748477 DOI: 10.3389/fmicb.2022.1005215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/07/2022] [Indexed: 07/30/2023] Open
Abstract
Salmonella enterica subsp. enterica (S.) serovar Cerro is rarely isolated from human clinical cases of salmonellosis but represents the most common serovar isolated from cattle without clinical signs of illness in the United States. In this study, using a large, diverse set of 316 isolates, we utilized genomic methods to further elucidate the evolutionary history of S. Cerro and to identify genomic features associated with its apparent virulence attenuation in humans. Phylogenetic analyses showed that within this polyphyletic serovar, 98.4% of isolates (311/316) represent a monophyletic clade within section Typhi and the remaining 1.6% of isolates (5/316) form a monophyletic clade within subspecies enterica Clade A1. Of the section Typhi S. Cerro isolates, 93.2% of isolates (290/311) clustered into a large clonal clade comprised of predominantly sequence type (ST) 367 cattle and environmental isolates, while the remaining 6.8% of isolates (21/311), primarily from human clinical sources, clustered outside of this clonal clade. A tip-dated phylogeny of S. Cerro ST367 identified two major clades (I and II), one of which overwhelmingly consisted of cattle isolates that share a most recent common ancestor that existed circa 1975. Gene presence/absence and rarefaction curve analyses suggested that the pangenome of section Typhi S. Cerro is open, potentially reflecting the gain/loss of prophage; human isolates contained the most open pangenome, while cattle isolates had the least open pangenome. Hypothetically disrupted coding sequences (HDCs) displayed clade-specific losses of intact speC and sopA virulence genes within the large clonal S. Cerro clade, while loss of intact vgrG, araH, and vapC occurred in all section Typhi S. Cerro isolates. Further phenotypic analysis suggested that the presence of a premature stop codon in speC does not abolish ornithine decarboxylase activity in S. Cerro, likely due to the activity of the second ornithine decarboxylase encoded by speF, which remained intact in all isolates. Overall, our study identifies specific genomic features associated with S. Cerro's infrequent isolation from humans and its apparent adaptation to cattle, which has broader implications for informing our understanding of the evolutionary events facilitating host adaptation in Salmonella.
Collapse
Affiliation(s)
- Alexa R. Cohn
- Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Renato H. Orsi
- Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Laura M. Carroll
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jingqiu Liao
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Martin Wiedmann
- Department of Food Science, Cornell University, Ithaca, NY, United States
| | - Rachel A. Cheng
- Department of Food Science, Cornell University, Ithaca, NY, United States
| |
Collapse
|
44
|
Mohakud NK, Panda RK, Patra SD, Sahu BR, Ghosh M, Kushwaha GS, Misra N, Suar M. Genome analysis and virulence gene expression profile of a multi drug resistant Salmonella enterica serovar Typhimurium ms202. Gut Pathog 2022; 14:28. [PMID: 35765034 PMCID: PMC9237969 DOI: 10.1186/s13099-022-00498-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/19/2022] [Indexed: 12/01/2022] Open
Abstract
Background In India, multi-drug resistance in Salmonella enterica serovar Typhimurium poses a significant health threat. Indeed, S. Typhimurium has remained unknown for a large portion of its genome associated with various physiological functions including mechanism of drug resistance and virulence. The whole-genome sequence of a Salmonella strain obtained from feces of a patient with gastroenteritis in Odisha, India, was analyzed for understanding the disease association and underlying virulence mechanisms. Results The de novo assembly yielded 17 contigs and showed 99.9% similarity to S. enterica sub sp enterica strain LT2 and S. enteric subsp salamae strain DSM 9220. S. Typhimurium ms202 strain constitutes six known Salmonella pathogenicity islands and nine different phages. The comparative interpretation of pathogenic islands displayed the genes contained in SPI-1 and SPI-2 to be highly conserved. We identified sit ABCD cluster regulatory cascade in SPI-1. Multiple antimicrobial resistance genes were identified that directly implies antibiotic-resistant phenotype. Notably, seven unique genes were identified as "acquired antibiotic resistance". These data suggest that virulence in S. enterica Typhimurium ms202 is associated with SPI-1 and SPI-2. Further, we found several virulent genes encoding SPI regions belonging to type III secretion systems (T3SS) of bacteria were significantly upregulated in ms202 compared to control LT2. Moreover, all these genes were significantly downregulated in S. enterica Typhimurium ms202 as compared to control LT2 on adding Mn2+ exogenously. Conclusions Our study raises a vital concern about the potential diffusion of a novel multi-drug resistant S. enterica Typhimurium ms202. It justifies this clinical pathogen to demonstrate a higher degree survival due to higher expression of virulent genes and enhanced ability of metallic ion acquisition. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-022-00498-w.
Collapse
Affiliation(s)
- Nirmal Kumar Mohakud
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, India.,Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, 751024, India
| | | | | | | | - Mrinmoy Ghosh
- KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar, 751024, India.
| | - Gajraj Singh Kushwaha
- KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar, 751024, India
| | - Namrata Misra
- KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar, 751024, India
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, India. .,KIIT-Technology Business Incubator (KIIT-TBI), KIIT University, Bhubaneswar, 751024, India.
| |
Collapse
|
45
|
Mechanisms for the Invasion and Dissemination of Salmonella. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2022; 2022:2655801. [PMID: 35722038 PMCID: PMC9203224 DOI: 10.1155/2022/2655801] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/15/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
Abstract
Salmonella enterica is a gastroenteric Gram-negative bacterium that can infect both humans and animals and causes millions of illnesses per year around the world. Salmonella infections usually occur after the consumption of contaminated food or water. Infections with Salmonella species can cause diseases ranging from enterocolitis to typhoid fever. Salmonella has developed multiple strategies to invade and establish a systemic infection in the host. Different cell types, including epithelial cells, macrophages, dendritic cells, and M cells, are important in the infection process of Salmonella. Dissemination throughout the body and colonization of remote organs are hallmarks of Salmonella infection. There are several routes for the dissemination of Salmonella typhimurium. This review summarizes the current understanding of the infection mechanisms of Salmonella. Additionally, different routes of Salmonella infection will be discussed. In this review, the strategies used by Salmonella enterica to establish persistent infection will be discussed. Understanding both the bacterial and host factors leading to the successful colonization of Salmonella enterica may enable the rational design of effective therapeutic strategies.
Collapse
|
46
|
Oueslati W, Rjeibi MR, Benyedem H, Mamlouk A, Souissi F, Selmi R, Ettriqui A. Salmonella Broiler Meat's Contamination in Tunisia: Prevalence, Serotypes, Antimicrobial Resistance and Molecular Characterization of Isolated Strains. Curr Microbiol 2022; 79:208. [PMID: 35639195 DOI: 10.1007/s00284-022-02900-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/09/2022] [Indexed: 11/03/2022]
Abstract
This study was conducted in north-eastern Tunisia to estimate the contamination prevalence of Salmonella in broilers' meat, to rank serotypes and to characterize the isolated multidrug-resistant (MDR) strains. A total number of 1288 meat samples were collected from 322 broiler batches; Salmonella isolates were identified by the alternative technique VIDAS Easy Salmonella. The susceptibility of Salmonella isolates was assessed against 21 antimicrobials using the disc diffusion method on Mueller-Hinton agar. Some antimicrobial resistance genes were identified using Polymerase Chain Reaction (PCR). The prevalence rates of Salmonella in the neck skin and the breast muscle contamination were estimated at 11.8% (38/322) and 0.9% (3/322), respectively. The prevalence rate of Salmonella in meat cutting parts contamination was estimated at 5.1% (33/644). Eight serotypes of Salmonella were identified, namely S. Enteritidis, S. Kentucky, S. Anatum, S. Infantis, S. Mbandaka, S. Zanzibar, S. Hadar and S. Agona. High rate of resistance was identified against amoxicillin (91.9%), nalidixic acid (83.8%), tetracycline (75.7%), streptomycin (73%), ciprofloxacin (70%), sulfamides (68.9%), cefalotin (68.9%), cefotaxim (67.6%) and cefoxitin (60.8%). The majority (90.5%; 67/74) of isolated strains was recognized as MDR. Nine MDR strains were identified as Extended-Spectrum β-Lactamase (ESBL) producers. The blaCTX-M gene was identified by PCR in all the nine ESBL strains. TetA, tetB and dfrA1 genes were amplified in 3.6% (2/56), 1.8% (1/56) and 19.3% (5/26) of tetracycline and trimethoprim-resistant strains, respectively. The integrase gene (class 2) was identified in only 8.1% (6/74) of the Salmonella-isolated strains. Our findings highlight the emergence of MDR Salmonella isolates in Tunisia.
Collapse
Affiliation(s)
- Walid Oueslati
- Laboratory of Management of Animal Production's Health and Quality, National School of Veterinary Medicine of Sidi Thabet, University Manouba (LR14AGR03), 2010, La Manouba, Tunisia. .,Department of Animal Production, National Agronomic Institute, University Carthage, 1054, Carthage, Tunisia.
| | - Mohamed Ridha Rjeibi
- Laboratory of Parasitology, National School of Veterinary Medicine of Sidi Thabet, University Manouba, 2010, La Manouba, Tunisia.,Laboratory of Parasitology, Veterinary Research Institute, University de Tunis El Manar, 1068, Tunis, Tunisia
| | - Hayet Benyedem
- Laboratory of Parasitology, National School of Veterinary Medicine of Sidi Thabet, University Manouba, 2010, La Manouba, Tunisia
| | - Aymen Mamlouk
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University Manouba, 2010, La Manouba, Tunisia
| | - Fatma Souissi
- Laboratory of Management of Animal Production's Health and Quality, National School of Veterinary Medicine of Sidi Thabet, University Manouba (LR14AGR03), 2010, La Manouba, Tunisia
| | - Rachid Selmi
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University Manouba, 2010, La Manouba, Tunisia
| | - Abdelfettah Ettriqui
- Laboratory of Management of Animal Production's Health and Quality, National School of Veterinary Medicine of Sidi Thabet, University Manouba (LR14AGR03), 2010, La Manouba, Tunisia
| |
Collapse
|
47
|
Walpole GFW, Pacheco J, Chauhan N, Clark J, Anderson KE, Abbas YM, Brabant-Kirwan D, Montaño-Rendón F, Liu Z, Zhu H, Brumell JH, Deiters A, Stephens LR, Hawkins PT, Hammond GRV, Grinstein S, Fairn GD. Kinase-independent synthesis of 3-phosphorylated phosphoinositides by a phosphotransferase. Nat Cell Biol 2022; 24:708-722. [PMID: 35484249 PMCID: PMC9107517 DOI: 10.1038/s41556-022-00895-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 03/08/2022] [Indexed: 01/10/2023]
Abstract
Despite their low abundance, phosphoinositides play a central role in membrane traffic and signalling. PtdIns(3,4,5)P3 and PtdIns(3,4)P2 are uniquely important, as they promote cell growth, survival and migration. Pathogenic organisms have developed means to subvert phosphoinositide metabolism to promote successful infection and their survival in host organisms. We demonstrate that PtdIns(3,4)P2 is a major product generated in host cells by the effectors of the enteropathogenic bacteria Salmonella and Shigella. Pharmacological, gene silencing and heterologous expression experiments revealed that, remarkably, the biosynthesis of PtdIns(3,4)P2 occurs independently of phosphoinositide 3-kinases. Instead, we found that the Salmonella effector SopB, heretofore believed to be a phosphatase, generates PtdIns(3,4)P2 de novo via a phosphotransferase/phosphoisomerase mechanism. Recombinant SopB is capable of generating PtdIns(3,4,5)P3 and PtdIns(3,4)P2 from PtdIns(4,5)P2 in a cell-free system. Through a remarkable instance of convergent evolution, bacterial effectors acquired the ability to synthesize 3-phosphorylated phosphoinositides by an ATP- and kinase-independent mechanism, thereby subverting host signalling to gain entry and even provoke oncogenic transformation.
Collapse
Affiliation(s)
- Glenn F W Walpole
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Pacheco
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neha Chauhan
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | - Yazan M Abbas
- Molecular Medicine Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Fernando Montaño-Rendón
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Zetao Liu
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Hongxian Zhu
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - John H Brumell
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sergio Grinstein
- Division of Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
48
|
Qasim M, Wrage M, Nüse B, Mattner J. Shigella Outer Membrane Vesicles as Promising Targets for Vaccination. Int J Mol Sci 2022; 23:994. [PMID: 35055181 PMCID: PMC8781765 DOI: 10.3390/ijms23020994] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/17/2022] Open
Abstract
The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell-cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.
Collapse
Affiliation(s)
- Muhammad Qasim
- Department of Microbiology, Kohat University of Science and Technology, Kohat 26000, Pakistan;
| | - Marius Wrage
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.W.); (B.N.)
| | - Björn Nüse
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.W.); (B.N.)
| | - Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.W.); (B.N.)
| |
Collapse
|
49
|
Tanui CK, Karanth S, Njage PM, Meng J, Pradhan AK. Machine learning-based predictive modeling to identify genotypic traits associated with Salmonella enterica disease endpoints in isolates from ground chicken. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2021.112701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
50
|
Prevalence, Risk Factors, Antimicrobial Resistance and Molecular Characterization of Salmonella in Northeast Tunisia Broiler Flocks. Vet Sci 2021; 9:vetsci9010012. [PMID: 35051096 PMCID: PMC8780282 DOI: 10.3390/vetsci9010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/27/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
This study was conducted in northeastern Tunisia to estimate both the prevalence and the risk factors of Salmonella in broiler flocks as well as to characterize the isolated multidrug-resistant (MDR) Salmonella strains. In the present study, a total number of 124 farms were sampled; Salmonella isolates were identified by the alternative technique VIDAS Easy Salmonella. The susceptibility of Salmonella isolates was assessed against 21 antimicrobials using the disk diffusion method on Mueller–Hinton agar using antimicrobial discs. Some antimicrobial resistance genes were identified using PCR. The prevalence rate of Salmonella infection, in the sampled farms, was estimated at 19.9% (64/322). Moreover, a total number of 13 different serotypes were identified. High rate of resistance was identified against nalidixic acid (82.85%), amoxicillin (81.25%), streptomycin (75%), and ciprofloxacin (75%). Alarming level of resistance to ertapenem (12.5%) was noticed. A total of 87.5% (56/64) of isolated strains were recognized as MDR. Three MDR strains were extended-spectrum β-lactamases (ESBL)-producers and three MDR strains were cephalosporinase-producers. The blaCTX-M gene was amplified in all the three ESBL strains. The qnrB gene was not amplified in fluoroquinolones-resistant strains. The tetA and tetB genes were amplified in 5% (2/40) and 2.5% (1/40) of tetracycline-resistant strains, respectively. The dfrA1 gene was amplified in five of the 20 trimethoprim-resistant strains. The mcr-1, mcr-2, mcr-3, mcr-4, and mcr-5 genes were not amplified in any of the phenotypically colistin-resistant strains. In terms of integrase genes int1 and int2, only gene class 2 was amplified in 11% (7/64) of analyzed strains. Risk factors, such as the poor level of cleaning and disinfection, the lack of antimicrobial treatment at the start of the breeding, and a crawl space duration lower than 15 days, were associated with high Salmonella infection in birds. These data should be considered when preparing salmonellosis control programs in Tunisian broiler flocks.
Collapse
|