1
|
Dang TV, Jang IS, Nguyen QH, Choi HS, Yu BJ, Kim MI. Signal-off colorimetric and signal-on fluorometric dual-mode aptasensor for ultrasensitive detection of Salmonella Typhimurium using graphitic carbon nitride. Food Chem 2025; 465:142176. [PMID: 39579407 DOI: 10.1016/j.foodchem.2024.142176] [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: 08/07/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Food safety is severely burdened by the prevalence of foodborne pathogens and the diseases they cause, necessitating the development of rapid, easy-to-use, highly sensitive, and reliable detection methods. Here, a signal-off colorimetric and signal-on fluorometric dual-mode detection method for Salmonella Typhimurium (S. typhimurium) was developed based on its unique interaction with aptamer DNA and graphitic carbon nitride (GCN). In the absence of a target Salmonella species, 6-carboxyfluorescein (FAM)-labeled aptamers are adsorbed on the surface of GCN primarily via a π-π interaction, resulting in reduced fluorescence of FAM through GCN-mediated quenching as well as improved peroxidase-like activity of GCN to generate intense blue color through facilitated electrostatic attraction between the negatively charged aptamer and positively charged 3,3',5,5'-tetramethylbenzidine (TMB) substrate. The introduction of S. typhimurium to the sample solution causes the detachment of the aptamer from GCN due to its higher affinity for S. typhimurium than GCN, thereby rapidly reducing the colorimetric signal and recovering the fluorescence. We successfully determined the number of S. typhimurium using this method in a remarkably short duration (10-30 min), highlighting its rapidity. The limit of detection values for S. typhimurium were as low as 8 and 3 CFU/mL when using colorimetric and fluorometric methods, respectively. Moreover, this method can be used to detect S. typhimurium spiked in real vegetable extract and milk with high reproducibility and reliability. This method serves as a convenient route to the rapid, sensitive, selective, and reliable detection of pathogens from complex food samples, with the potential to replace conventional yet laborious methods currently in use.
Collapse
Affiliation(s)
- Thinh Viet Dang
- Department of BioNano Technology, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam, Gyeonggi 13120, Republic of Korea
| | - In Seung Jang
- Low-Carbon Transition R&D Department, Research Institute of Sustainable Development Technology, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Republic of Korea
| | - Quynh Huong Nguyen
- Department of BioNano Technology, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam, Gyeonggi 13120, Republic of Korea
| | - Hyeun Seok Choi
- Regional Industrial Innovation Department, Research Institute of Sustainable Development Technology, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Republic of Korea
| | - Byung Jo Yu
- Low-Carbon Transition R&D Department, Research Institute of Sustainable Development Technology, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Republic of Korea.
| | - Moon Il Kim
- Department of BioNano Technology, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam, Gyeonggi 13120, Republic of Korea.
| |
Collapse
|
2
|
Jyoti, Castillo AR, Jurado-Sánchez B, Pumera M, Escarpa A. Active Quantum Biomaterials-Enhanced Microrobots for Food Safety. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404248. [PMID: 39449211 DOI: 10.1002/smll.202404248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/22/2024] [Indexed: 10/26/2024]
Abstract
Timely disruptive tools for the detection of pathogens in foods are needed to face global health and economic challenges. Herein, the utilization of quantum biomaterials-enhanced microrobots (QBEMRs) as autonomous mobile sensors designed for the precise detection of endotoxins originating from Salmonella enterica (S. enterica) as an indicator species for food-borne contamination globally is presented. A fluorescent molecule-labeled affinity peptide functions as a specific probe, is quenched upon binding to the surface of QBEMRs. Owing to its selective affinity for endotoxin, in the presence of S. enterica the fluorescence is restored and easy to observe and quantifies optical color change to indicate the presence of Salmonella. The devised approach is designed to achieve highly sensitive detection of the S. enterica serovar Typhimurium endotoxin with exquisite selectivity through the utilization of QBEMRs. Notably, no fluorescence signal is observed in the presence of endotoxins bearing similar structural characteristics, highlighting the selectivity of the approach during food sample analysis. Technically, the strategy is implemented in microplate readers to extend microrobots-based approaches to the routine laboratory. This new platform can provide fast and anticipated results in food safety.
Collapse
Affiliation(s)
- Jyoti
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
- Future Energy and Innovation Laboratory, Central European Institute of Technology, Brno University of Technology (CEITEC-BUT), Brno, 61200, Czech Republic
| | - Alberto-Rodríguez Castillo
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
| | - Beatriz Jurado-Sánchez
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
- Chemical Research Institute "Andres M. Del Río,", Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
| | - Martin Pumera
- Future Energy and Innovation Laboratory, Central European Institute of Technology, Brno University of Technology (CEITEC-BUT), Brno, 61200, Czech Republic
- Advanced Nanorobots & Multiscale Robotics Laboratory, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, 17. Listopadu 2172/15, Ostrava, 70800, Czech Republic
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Alberto Escarpa
- Department of Analytical Chemistry, Physical Chemistry, and Chemical Engineering, Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
- Chemical Research Institute "Andres M. Del Río,", Universidad de Alcala, Alcala de Henares, Madrid, E-28802, Spain
| |
Collapse
|
3
|
Bisola Bello A, Olamilekan Adesola R, Idris I, Yawson Scott G, Alfa S, Akinfemi Ajibade F. Combatting extensively drug-resistant Salmonella: a global perspective on outbreaks, impacts, and control strategies. Pathog Glob Health 2024:1-15. [PMID: 39508610 DOI: 10.1080/20477724.2024.2416864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Antibiotic resistance in typhoid fever poses a critical public health problem due to the emergence of extensively drug-resistant (XDR) Salmonella, resulting in prolonged illness and treatment failure. Salmonella enterica serovar Typhi is the most predominant among all serotypes and can acquire resistance. The emergence of XDR Salmonella in various regions globally, particularly Pakistan, presents a concerning trend. However, limited data availability impedes a comprehensive understanding of the outbreaks and hinders the development of real-time solutions. Here, we have provided an updated overview of the current outbreaks of XDR Salmonella in epidemic and endemic regions. Treatments of XDR Salmonella infections are challenging, as there are records of treatment failure in humans and animals. However, intensive prevention techniques can be implemented pending the advent of novel antibiotics. Emphasis on antimicrobial stewardship and frequent surveillance of the pathogen should be made to keep track of potential outbreaks in both human and animal populations. Although progress is being made to combat XDR Salmonella within some regions, a unified and efficient effort on an international scale is required to curtail the XDR outbreak before it escalates and leads us back to the pre-antibiotic era.
Collapse
Affiliation(s)
- Aisha Bisola Bello
- Department of Biological Science, School of Applied and Natural Sciences, Federal Polytechnic Bida, Bida, Nigeria
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ahmadu Bello University, Zaria, Nigeria
| | - Ridwan Olamilekan Adesola
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ibrahim Idris
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Godfred Yawson Scott
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Suleiman Alfa
- Department of Biological Science, School of Applied and Natural Sciences, Federal Polytechnic Bida, Bida, Nigeria
| | - Favour Akinfemi Ajibade
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
4
|
Kim S, Lee YH. The Salmonella enterica EnvE is an Outer Membrane Lipoprotein and Its Gene Expression Leads to Transcriptional Repression of the Virulence Gene msgA. J Microbiol 2024; 62:1013-1022. [PMID: 39546166 DOI: 10.1007/s12275-024-00183-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/18/2024] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
The envE gene of Salmonella enterica serovar Typhimurium is encoded within Salmonella Pathogenicity Island-11 (SPI-11) and is located immediately upstream of the virulence gene msgA (macrophage survival gene A) in the same transcriptional orientation. To date, the characteristics and roles of envE remain largely unexplored. In this study, we show that EnvE, a predicted lipoprotein, is localized on the outer membrane using sucrose gradient ultracentrifugation. Under oxidative stress conditions, envE transcription is suppressed, while msgA transcription is induced, indicating an inverse correlation between the mRNA levels of the two neighboring genes. Importantly, inactivation of envE leads to constitutive transcription of msgA regardless of the presence of oxidative stress. Moreover, trans-complementation of the envE mutant with a plasmid-borne envE fails to prevent the induction of msgA transcription, suggesting that envE functions as a cis-regulatory element rather than a trans-acting factor. We further show that both inactivation and complementation of envE confer wild-type levels of resistance to oxidative stress by ensuring the expression of msgA. Our data suggest that the S. enterica envE gene encodes an outer membrane lipoprotein, and its transcription represses msgA expression in a cis-acting manner, probably by transcriptional interference, although the exact molecular details are yet unclear.
Collapse
Affiliation(s)
- Sinyeon Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
- MThera Pharma Co., Seoul, 07793, Republic of Korea
| | - Yong Heon Lee
- Department of Biomedical Laboratory Science, Dongseo University, Busan, 47011, Republic of Korea.
| |
Collapse
|
5
|
Chen R, Yang L, Pajor MS, Wiedmann M, Orsi RH. Salmonella associated with agricultural animals exhibit diverse evolutionary rates and show evidence of recent clonal expansion. mBio 2024; 15:e0191324. [PMID: 39287448 PMCID: PMC11492988 DOI: 10.1128/mbio.01913-24] [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: 07/22/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
Most foodborne salmonellosis outbreaks are linked to agricultural animal products with a few serovars accounting for most Salmonella isolated from specific animal products, suggesting an adaptation to the corresponding animal hosts and their respective environments. Here, we utilized whole-genome sequence (WGS) data to analyze the evolution and population genetics of seven serovars frequently isolated from ground beef (Montevideo, Cerro, and Dublin), chicken (Kentucky, Infantis, and Enteritidis), and turkey (Reading) in the United States. In addition, publicly available metadata were used to characterize major clades within each serovar with regard to public health significance. Except for Dublin, all serovars were polyphyletic, comprising 2-6 phylogenetic groups. Further partitioning of the phylogenies identified 25 major clades, including 12 associated with animal or environmental niches. These 12 clades differed in evolutionary parameters (e.g., substitution rates) as well as public health relevant characteristics (e.g., association with human illness, antimicrobial resistance). Overall, our results highlight several critical trends: (i) the Salmonella generation time appears to be more dependent on source than serovar and (ii) all serovars contain clades and sub-clades that are estimated to have emerged after the year 1940 and that are enriched for isolates associated with humans, agricultural animals, antimicrobial resistance (AMR), and/or specific geographical regions. These findings suggest that serotyping alone does not provide enough resolution to differentiate isolates that may have evolved independently, present distinct geographic distribution and host association, and possibly have distinct public health significance. IMPORTANCE Non-typhoidal Salmonella are major foodborne bacterial pathogens estimated to cause more than one million illnesses, thousands of hospitalizations, and hundreds of deaths annually in the United States. More than 70% of Salmonella outbreaks in the United States have been associated with agricultural animals. Certain serovars include persistent strains that have repeatedly contaminated beef, chicken, and turkey, causing outbreaks and sporadic cases over many years. These persistent strains represent a particular challenge to public health, as they are genetically clonal and widespread, making it difficult to differentiate distinct outbreak and contamination events using whole-genome sequence (WGS)-based subtyping methods (e.g., core genome allelic typing). Our results indicate that a phylogenetic approach is needed to investigate persistent strains and suggest that the association between a Salmonella serovar and an agricultural animal is driven by the expansion of clonal subtypes that likely became adapted to specific animals and associated environments.
Collapse
Affiliation(s)
- Ruixi Chen
- Department of Food
Science, Cornell University,
Ithaca, New York, USA
| | - Linghuan Yang
- Department of Food
Science, Cornell University,
Ithaca, New York, USA
| | | | - Martin Wiedmann
- Department of Food
Science, Cornell University,
Ithaca, New York, USA
| | - Renato H. Orsi
- Department of Food
Science, Cornell University,
Ithaca, New York, USA
| |
Collapse
|
6
|
Delyea CJ, Forster MD, Luo S, Dubrule BE, Julien O, Bhavsar AP. The Salmonella Effector SspH2 Facilitates Spatially Selective Ubiquitination of NOD1 to Enhance Inflammatory Signaling. Biochemistry 2024; 63:2266-2279. [PMID: 39189508 PMCID: PMC11412229 DOI: 10.1021/acs.biochem.4c00380] [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: 06/28/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
As part of its pathogenesis, Salmonella enterica serovar Typhimurium delivers effector proteins into host cells. One effector is SspH2, a member of the so-called novel E3 ubiquitin ligase family, that interacts with and enhances, NOD1 pro-inflammatory signaling, though the underlying mechanisms are unclear. Here, we report that SspH2 interacts with multiple members of the NLRC family to enhance pro-inflammatory signaling by targeted ubiquitination. We show that SspH2 modulates host innate immunity by interacting with both NOD1 and NOD2 in mammalian epithelial cell culture via the NF-κB pathway. Moreover, purified SspH2 and NOD1 directly interact, where NOD1 potentiates SspH2 E3 ubiquitin ligase activity. Mass spectrometry and mutational analyses identified four key lysine residues in NOD1 that are required for its enhanced activation by SspH2, but not its basal activity. These critical lysine residues are positioned in the same region of NOD1 and define a surface on the receptor that appears to be targeted by SspH2. Overall, this work provides evidence for post-translational modification of NOD1 by ubiquitin and uncovers a unique mechanism of spatially selective ubiquitination to enhance the activation of an archetypal NLR.
Collapse
Affiliation(s)
- Cole J. Delyea
- Department
of Medical Microbiology and Immunology, Faculty of Medicine &
Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Malcolm D. Forster
- Department
of Medical Microbiology and Immunology, Faculty of Medicine &
Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Shu Luo
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Bradley E. Dubrule
- Department
of Medical Microbiology and Immunology, Faculty of Medicine &
Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Amit P. Bhavsar
- Department
of Medical Microbiology and Immunology, Faculty of Medicine &
Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
7
|
Zhuang H, Wang R, Qi Y, Liu Y, Xiong H, Yao J. Nanocoated bacteria with H 2S generation-triggered self-amplified photothermal and photodynamic effect for breast cancer therapy. J Control Release 2024; 373:507-519. [PMID: 39025267 DOI: 10.1016/j.jconrel.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Phototherapy utilizing bacterial carriers has demonstrated efficacy in anti-tumor therapy, while the poor delivery of phototherapeutic agents and immunogenicity of microbial substances remain problematic. Herein, we develop a nanocoated bacterial delivery system (IF-S.T) that in situ forms the efficient photothermal agents via biomineralization and improves the intracellular oxygenation, thus triggering the self-enhanced photothermal therapy (PTT) and photodynamic therapy (PDT) on tumor. We densely coat self-assembled IF (ICG-Fe2+) nanocomplex onto the surface of LT2, weakly virulent strain of Salmonella typhimurium (S.T), by bioadaptive nanocoating techniques, masking bacterial virulence factors and reducing the potential immune adverse effects. Upon penetrating into the tumor environment, IF-S.T responds to H2O2 to trigger the removal of the IF coating, where S.T produces excess hydrogen sulfide (H2S). H2S reacts with Fe2+, yielding ferrous sulfide (FeS) for PTT, and inhibits mitochondrial respiration to enhance tumor cell oxygenation for PDT. Consequently, IF-S.T plus laser irradiation exhibits direct tumor cells killing and elicits robust antitumor immune responses, leading to the complete tumor elimination. Thus, IF-S.T represents a promising platform for effective tumor delivery of photoactive agents for improved PTT/PDT efficacy.
Collapse
Affiliation(s)
- He Zhuang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Rui Wang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yao Qi
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yufei Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Hui Xiong
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Jing Yao
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| |
Collapse
|
8
|
Li H, Xu H, Shi X, Zhao C, Li J, Wang J. Colorimetry/fluorescence dual-mode detection of Salmonella typhimurium based on a "three-in-one" nanohybrid with high oxidase-like activity for AIEgen. Food Chem 2024; 449:139220. [PMID: 38579657 DOI: 10.1016/j.foodchem.2024.139220] [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: 12/03/2023] [Revised: 03/03/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
A colorimetry/fluorescence dual-mode assay based on the aptamer-functionalized magnetic covalent organic framework-supported CuO and Au NPs (MCOF-CuO/Au@apt) was developed for Salmonella typhimurium (S. typhimurium) biosensing. The nanohybrid combined three functions in one: good magnetic separation characteristic, excellent oxidase-mimic activity for tetrap-aminophenylethylene (TPE-4A), and target recognition capability. The attachment of MCOF-CuO/Au@apt onto the surface of S. typhimurium resulted in a significant reduction in the oxidase-mimicking activity of the nanohybrid, which could generate dual-signal of colorimetry and fluorescence through the catalytic oxidation of TPE-4A. Based on this, S. typhimurium could be specifically detected in the linear ranges of 102- 106 CFU·mL-1 and 101- 106 CFU·mL-1, with LODs of 7.6 and 2.1 CFU·mL-1, respectively in colorimetry/fluorescence modes. Moreover, the smartphone and linear discrimination analysis-based system could be used for on-site and portable testing. In addition, this platform showed applicability in detecting S. typhimurium in milk, egg liquid and chicken samples.
Collapse
Affiliation(s)
- Hang Li
- School of Public Health, Jilin University, Changchun 130021, China
| | - Hui Xu
- School of Public Health, Jilin University, Changchun 130021, China
| | - Xuening Shi
- School of Public Health, Jilin University, Changchun 130021, China
| | - Chao Zhao
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Juan Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Juan Wang
- School of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
McDonald K, Rodriguez A, Muthukrishnan G. Humanized Mouse Models of Bacterial Infections. Antibiotics (Basel) 2024; 13:640. [PMID: 39061322 PMCID: PMC11273811 DOI: 10.3390/antibiotics13070640] [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: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Bacterial infections continue to represent a significant healthcare burden worldwide, causing considerable mortality and morbidity every year. The emergence of multidrug-resistant bacterial strains continues to rise, posing serious risks to controlling global disease outbreaks. To develop novel and more effective treatment and vaccination programs, there is a need for clinically relevant small animal models. Since multiple bacterial species have human-specific tropism for numerous virulence factors and toxins, conventional mouse models do not fully represent human disease. Several human disease characteristic phenotypes, such as lung granulomas in the case of Mycobacterium tuberculosis infections, are absent in standard mouse models. Alternatively, certain pathogens, such as Salmonella enterica serovar typhi and Staphylococcus aureus, can be well tolerated in mice and cleared quickly. To address this, multiple groups have developed humanized mouse models and observed enhanced susceptibility to infection and a more faithful recapitulation of human disease. In the last two decades, multiple humanized mouse models have been developed to attempt to recapitulate the human immune system in a small animal model. In this review, we first discuss the history of immunodeficient mice that has enabled the engraftment of human tissue and the engraftment methods currently used in the field. We then highlight how humanized mouse models successfully uncovered critical human immune responses to various bacterial infections, including Salmonella enterica serovar Typhi, Mycobacterium tuberculosis, and Staphylococcus aureus.
Collapse
Affiliation(s)
- Katya McDonald
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Adryiana Rodriguez
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
10
|
Mylona E, Pham Thanh D, Keane JA, Dongol S, Basnyat B, Dolecek C, Voong Vinh P, Tran Vu Thieu N, Nguyen Thi Nguyen T, Karkey A, Baker S. A retrospective investigation of the population structure and geospatial distribution of Salmonella Paratyphi A in Kathmandu, Nepal. PLoS Negl Trop Dis 2024; 18:e0011864. [PMID: 38889189 PMCID: PMC11216570 DOI: 10.1371/journal.pntd.0011864] [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: 12/14/2023] [Revised: 07/01/2024] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
Salmonella Paratyphi A, one of the major etiologic agents of enteric fever, has increased in prevalence in recent decades in certain endemic regions in comparison to S. Typhi, the most prevalent cause of enteric fever. Despite this increase, data on the prevalence and molecular epidemiology of S. Paratyphi A remain generally scarce. Here, we analysed the whole genome sequences of 216 S. Paratyphi A isolates originating from Kathmandu, Nepal between 2005 and 2014, of which 200 were from patients with acute enteric fever and 16 from the gallbladder of people with suspected chronic carriage. By exploiting the recently developed genotyping framework for S. Paratyphi A (Paratype), we identified several genotypes circulating in Kathmandu. Notably, we observed an unusual clonal expansion of genotype 2.4.3 over a four-year period that spread geographically and systematically replaced other genotypes. This rapid genotype replacement is hypothesised to have been driven by both reduced susceptibility to fluoroquinolones and genetic changes to virulence factors, such as functional and structural genes encoding the type 3 secretion systems. Finally, we show that person-to-person is likely the most common mode of transmission and chronic carriers seem to play a limited role in maintaining disease circulation.
Collapse
Affiliation(s)
- Elli Mylona
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Duy Pham Thanh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Program, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Jacqueline A. Keane
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Sabina Dongol
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Buddha Basnyat
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Christiane Dolecek
- The Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Phat Voong Vinh
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Program, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Nga Tran Vu Thieu
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Program, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - To Nguyen Thi Nguyen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Abhilasha Karkey
- The Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, United Kingdom
- IAVI Human Immunology Laboratory, Chelsea and Westminster Hospital, London, United Kingdom
| |
Collapse
|
11
|
Wang BX, Leshchiner D, Luo L, Tuncel M, Hokamp K, Hinton JCD, Monack DM. High-throughput fitness experiments reveal specific vulnerabilities of human-adapted Salmonella during stress and infection. Nat Genet 2024; 56:1288-1299. [PMID: 38831009 PMCID: PMC11176087 DOI: 10.1038/s41588-024-01779-7] [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: 09/12/2023] [Accepted: 04/25/2024] [Indexed: 06/05/2024]
Abstract
Salmonella enterica is comprised of genetically distinct 'serovars' that together provide an intriguing model for exploring the genetic basis of pathogen evolution. Although the genomes of numerous Salmonella isolates with broad variations in host range and human disease manifestations have been sequenced, the functional links between genetic and phenotypic differences among these serovars remain poorly understood. Here, we conduct high-throughput functional genomics on both generalist (Typhimurium) and human-restricted (Typhi and Paratyphi A) Salmonella at unprecedented scale in the study of this enteric pathogen. Using a comprehensive systems biology approach, we identify gene networks with serovar-specific fitness effects across 25 host-associated stresses encountered at key stages of human infection. By experimentally perturbing these networks, we characterize previously undescribed pseudogenes in human-adapted Salmonella. Overall, this work highlights specific vulnerabilities encoded within human-restricted Salmonella that are linked to the degradation of their genomes, shedding light into the evolution of this enteric pathogen.
Collapse
Affiliation(s)
- Benjamin X Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Lijuan Luo
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Miles Tuncel
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Karsten Hokamp
- Department of Genetics, School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Jay C D Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Denise M Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
12
|
Yang Y, Wang P, Qaidi SE, Hardwidge PR, Huang J, Zhu G. Loss to gain: pseudogenes in microorganisms, focusing on eubacteria, and their biological significance. Appl Microbiol Biotechnol 2024; 108:328. [PMID: 38717672 PMCID: PMC11078800 DOI: 10.1007/s00253-023-12971-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 05/12/2024]
Abstract
Pseudogenes are defined as "non-functional" copies of corresponding parent genes. The cognition of pseudogenes continues to be refreshed through accumulating and updating research findings. Previous studies have predominantly focused on mammals, but pseudogenes have received relatively less attention in the field of microbiology. Given the increasing recognition on the importance of pseudogenes, in this review, we focus on several aspects of microorganism pseudogenes, including their classification and characteristics, their generation and fate, their identification, their abundance and distribution, their impact on virulence, their ability to recombine with functional genes, the extent to which some pseudogenes are transcribed and translated, and the relationship between pseudogenes and viruses. By summarizing and organizing the latest research progress, this review will provide a comprehensive perspective and improved understanding on pseudogenes in microorganisms. KEY POINTS: • Concept, classification and characteristics, identification and databases, content, and distribution of microbial pseudogenes are presented. • How pseudogenization contribute to pathogen virulence is highlighted. • Pseudogenes with potential functions in microorganisms are discussed.
Collapse
Affiliation(s)
- Yi Yang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Joint Laboratory of International Cooperation On Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, China
| | - Pengzhi Wang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Joint Laboratory of International Cooperation On Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, China
| | - Samir El Qaidi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Jinlin Huang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Jiangsu Key Lab of Zoonosis, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
- College of Bioscience and Biotechnology, Yangzhou University, 12 East Wenhui Road Yangzhou, Jiangsu, 225009, China.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Joint Laboratory of International Cooperation On Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, China.
| |
Collapse
|
13
|
Chen Y, Liu L, Guo Y, Chu J, Wang B, Sui Y, Wei H, Hao H, Huang L, Cheng G. Distribution and genetic characterization of fluoroquinolone resistance gene qnr among Salmonella strains from chicken in China. Microbiol Spectr 2024; 12:e0300023. [PMID: 38411972 PMCID: PMC10986518 DOI: 10.1128/spectrum.03000-23] [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: 08/02/2023] [Accepted: 11/06/2023] [Indexed: 02/28/2024] Open
Abstract
The prevalence and dissemination of the plasmid-mediated fluoroquinolone (FQ) resistance gene qnr in Salmonella are considered serious public health concerns worldwide. So far, no comprehensive large-scale studies have focused on the prevalence and genetic characteristics of the qnr gene in Salmonella isolated from chickens. Herein, this study aimed to investigate the prevalence, antimicrobial resistance (AMR) patterns, and molecular characteristics of chicken-originated qnr-positive Salmonella strains from chicken farms, slaughterhouses, and markets in 12 provinces of China in 2020-2021. The overall prevalence of the qnr gene was 21.13% (56/265), with the highest prevalence in markets (36.11%, 26/72), followed in farms (17.95%, 21/117), and slaughterhouses (10.53%, 9/76). Only the qnrS and qnrB genes were detected, and the prevalence rate of the qnrS gene (19.25%, 51/265) was higher than that of the qnrB gene (1.89%, 5/265). Whole genome sequencing identified 37 distinct AMR genes and 15 plasmid replicons, and the most frequent mutation in quinolone resistance determining regions was parC (T57S; 91.49%, 43/47). Meanwhile, four different qnrS and two qnrB genetic environments were discovered among 47 qnr-positive Salmonella strains. In total, 21.28% (10/47) of the strains were capable of conjugative transfer, and all were qnrS1-positive strains, with the majority of transferable plasmids being IncHI2 types (n = 4). Overall, the prevalence of qnr-positive Salmonella strains from chickens in China and their carriage of multiple resistance and virulence genes and transferable plasmids is a major concern, which calls for continuous surveillance of qnr-positive Salmonella and the development of measures to control its prevalence and transmission.IMPORTANCESalmonella is a common foodborne pathogen responsible for 155,000 deaths annually worldwide. Fluoroquinolones (FQs) are used as first-line drugs for the treatment of Salmonella infections in several countries and regions. However, the emergence and increasing prevalence of the FQ-resistant gene qnr in Salmonella isolated from chickens have been widely reported. Gaining insight into the genetic mechanisms of AMR genes in chicken could lead to the development of preventive measures to control and reduce the risk of drug resistance. In this study, we identified qnr-positive Salmonellae isolated from chickens in different regions of China and their AMR patterns and genome-wide characteristics, providing a theoretical basis for further control of their prevalence and transmission.
Collapse
Affiliation(s)
- Yang Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lihui Liu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yali Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jinhua Chu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Bangjuan Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yuxin Sui
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hanqi Wei
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Haihong Hao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guyue Cheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
14
|
Marchant P, Vivanco E, Silva A, Nevermann J, Fuentes I, Barrera B, Otero C, Calderón IL, Gil F, Fuentes JA. β-lactam-induced OMV release promotes polymyxin tolerance in Salmonella enterica sv. Typhi. Front Microbiol 2024; 15:1389663. [PMID: 38591031 PMCID: PMC10999688 DOI: 10.3389/fmicb.2024.1389663] [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: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
The rise of multidrug-resistant bacteria is a global concern, leading to a renewed reliance on older antibiotics like polymyxins as a last resort. Polymyxins, cationic cyclic peptides synthesized nonribosomally, feature a hydrophobic acyl tail and positively charged residues. Their antimicrobial mechanism involves initial interaction with Gram-negative bacterial outer-membrane components through polar and hydrophobic interactions. Outer membrane vesicles (OMVs), nano-sized proteoliposomes secreted from the outer membrane of Gram-negative bacteria, play a crucial role in tolerating harmful molecules, including cationic peptides such as polymyxins. Existing literature has documented environmental changes' impact on modulating OMV properties in Salmonella Typhimurium. However, less information exists regarding OMV production and characteristics in Salmonella Typhi. A previous study in our laboratory showed that S. Typhi ΔmrcB, a mutant associated with penicillin-binding protein (PBP, a β-lactam antibiotic target), exhibited hypervesiculation. Consequently, this study investigated the potential impact of β-lactam antibiotics on promoting polymyxin tolerance via OMVs in S. Typhi. Our results demonstrated that sub-lethal doses of β-lactams increased bacterial survival against polymyxin B in S. Typhi. This phenomenon stems from β-lactam antibiotics inducing hypervesiculation of OMVs with higher affinity for polymyxin B, capturing and diminishing its biologically effective concentration. These findings suggest that β-lactam antibiotic use may inadvertently contribute to decreased polymyxin effectivity against S. Typhi or other Gram-negative bacteria, complicating the effective treatment of infections caused by these pathogens. This study emphasizes the importance of evaluating the influence of β-lactam antibiotics on the interaction between OMVs and other antimicrobial agents.
Collapse
Affiliation(s)
- Pedro Marchant
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Erika Vivanco
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Andrés Silva
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Jan Nevermann
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Ignacio Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Boris Barrera
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Iván L. Calderón
- Laboratorio de RNAs Bacterianos, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Fernando Gil
- Microbiota-Host Interactions and Clostridia Research Group, Universidad Andres Bello, Santiago, Chile
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
15
|
Cohen E, Azriel S, Auster O, Gal A, Mikhlin S, Crauwels S, Rahav G, Gal-Mor O. A new Salmonella enterica serovar that was isolated from a wild sparrow presents a distinct genetic, metabolic and virulence profile. Microbes Infect 2024; 26:105249. [PMID: 37956735 DOI: 10.1016/j.micinf.2023.105249] [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: 08/27/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Salmonella enterica is a ubiquitous and clinically-important bacterial pathogen, able to infect and cause different diseases in a wide range of hosts. Here, we report the isolation and characterization of a new S. enterica serovar (13,23:i:-; S. Tirat-Zvi), belonging to the Havana supper-lineage that was isolated from a wild house sparrow (Passer domesticus) in Israel. Whole genome sequencing and complete assembly of its genome indicated a plasmid-free, 4.7 Mb genome that carries the Salmonella pathogenicity islands 1-6, 9, 19 and an integrative and conjugative element (ICE), encoding arsenic resistance genes. Phenotypically, S. Tirat-Zvi isolate TZ282 was motile, readily formed biofilm, more versatile in carbon source utilization than S. Typhimurium and highly tolerant to arsenic, but impaired in host cell invasion. In-vivo infection studies indicated that while S. Tirat-Zvi was able to infect and cause an acute inflammatory enterocolitis in young chicks, it was compromised in mice colonization and did not cause an inflammatory colitis in mice compared to S. Typhimurium. We suggest that these phenotypes reflect the distinctive ecological niche of this new serovar and its evolutionary adaptation to passerine birds, as a permissive host. Moreover, these results further illuminate the genetic, phenotypic and ecological diversity of S. enterica pathovars.
Collapse
Affiliation(s)
- Emiliano Cohen
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Shalevet Azriel
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Oren Auster
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Adiv Gal
- Faculty of Sciences, Kibbutzim College, Tel-Aviv, Israel
| | | | - Sam Crauwels
- Laboratory for Process Microbial Ecology and Bioinspirational Management (PME&BIM), Centre of Microbial and Plant Genetics (CMPG), Department of Microbial and Molecular Systems (M2S), KU Leuven, Leuven, Belgium
| | - Galia Rahav
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Aguirre-Sanchez JR, Vega-Lopez IF, Castro Del Campo N, Medrano-Felix JA, Martínez-Urtaza J, Chaidez-Quiroz C. Genomic-wide analysis approach revealed genomic similarity for environmental Mexican S. Oranienburg genomes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:956-967. [PMID: 36946386 DOI: 10.1080/09603123.2023.2191312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
As the human population grows, an increase in food trade is needed. This elevates the risk of epidemiological outbreaks. One of the prevalent pathogens associated with food production in Mexico has been Salmonella Oranienburg. Effective surveillance systems require microbial genetic knowledge. The objective of this work is to describe the genetic composition of Mexican S. Oranienburg genomes. For that, 53 strains from different environmental sources were isolated and sequenced. Additionally, 109 S. Oranienburg genomes were downloaded. Bioinformatic analyses were used to explore the clonal complex and genomic relatedness. A major clonal group formed by ST23 was identified comprising four STs. 202 SNPs were found the maximum difference among isolates. Virulence genes for host invasion and colonization as rpoS, fimbria type 1, and, T3SS were found common for all isolates. This study suggests that Mexican S. Oranienburg strains are potential pathogens circulating continuously in the region between host and non-host environments.
Collapse
Affiliation(s)
- J R Aguirre-Sanchez
- Inocuidad Alimentaria, Centro de Investigación en Alimentación y Desarrollo, Culiacán, México
| | - I F Vega-Lopez
- Laboratorio en Ciencia y Mineria de Datos, Parque de Innovación Tecnológica de la Universidad Autónoma de Sinaloa, Culiacán, México
| | - N Castro Del Campo
- Inocuidad Alimentaria, Centro de Investigación en Alimentación y Desarrollo, Culiacán, México
| | - J A Medrano-Felix
- Inocuidad Alimentaria, Investigadoras e Investigadores por México-Centro de Investigación en Alimentación y Desarrollo A.C, Culiacán, México
| | - J Martínez-Urtaza
- Departement of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Chaidez-Quiroz
- Inocuidad Alimentaria, Centro de Investigación en Alimentación y Desarrollo, Culiacán, México
| |
Collapse
|
17
|
Benevides VP, Saraiva MMS, Nascimento CF, Delgado-Suárez EJ, Oliveira CJB, Silva SR, Miranda VFO, Christensen H, Olsen JE, Berchieri Junior A. Genomic Features and Phylogenetic Analysis of Antimicrobial-Resistant Salmonella Mbandaka ST413 Strains. Microorganisms 2024; 12:312. [PMID: 38399716 PMCID: PMC10893270 DOI: 10.3390/microorganisms12020312] [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: 12/29/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, Salmonella enterica subsp. enterica serovar Mbandaka (S. Mbandaka) has been increasingly isolated from laying hens and shell eggs around the world. Moreover, this serovar has been identified as the causative agent of several salmonellosis outbreaks in humans. Surprisingly, little is known about the characteristics of this emerging serovar, and therefore, we investigated antimicrobial resistance, virulence, and prophage genes of six selected Brazilian strains of Salmonella Mbandaka using Whole Genome Sequencing (WGS). Multi-locus sequence typing revealed that the tested strains belong to Sequence Type 413 (ST413), which has been linked to recent multi-country salmonellosis outbreaks in Europe. A total of nine resistance genes were detected, and the most frequent ones were aac(6')-Iaa, sul1, qacE, blaOXA-129, tet(B), and aadA1. A point mutation in ParC at the 57th position (threonine → serine) associated with quinolone resistance was present in all investigated genomes. A 112,960 bp IncHI2A plasmid was mapped in 4/6 strains. This plasmid harboured tetracycline (tetACDR) and mercury (mer) resistance genes, genes contributing to conjugative transfer, and genes involved in plasmid maintenance. Most strains (four/six) carried Salmonella genomic island 1 (SGI1). All S. Mbandaka genomes carried seven pathogenicity islands (SPIs) involved in intracellular survival and virulence: SPIs 1-5, 9, and C63PI. The virulence genes csgC, fimY, tcfA, sscA, (two/six), and ssaS (one/six) were absent in some of the genomes; conversely, fimA, prgH, and mgtC were present in all of them. Five Salmonella bacteriophage sequences (with homology to Escherichia phage phiV10, Enterobacteria phage Fels-2, Enterobacteria phage HK542, Enterobacteria phage ST64T, Salmonella phage SW9) were identified, with protein counts between 31 and 54, genome lengths of 24.7 bp and 47.7 bp, and average GC content of 51.25%. In the phylogenetic analysis, the genomes of strains isolated from poultry in Brazil clustered into well-supported clades with a heterogeneous distribution, primarily associated with strains isolated from humans and food. The phylogenetic relationship of Brazilian S. Mbandaka suggests the presence of strains with high epidemiological significance and the potential to be linked to foodborne outbreaks. Overall, our results show that isolated strains of S. Mbandaka are multidrug-resistant and encode a rather conserved virulence machinery, which is an epidemiological hallmark of Salmonella strains that have successfully disseminated both regionally and globally.
Collapse
Affiliation(s)
- Valdinete P Benevides
- Postgraduate Program in Agricultural Microbiology, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Mauro M S Saraiva
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Camila F Nascimento
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
| | - Enrique J Delgado-Suárez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Celso J B Oliveira
- Center for Agricultural Sciences, Department of Animal Science, Federal University of Paraiba (CCA/UFPB), Areia 58051-900, Brazil
- Global One Health Initiative (GOHi), The Ohio State University, Columbus, OH 43210, USA
| | - Saura R Silva
- Laboratory of Plant Systematics, Department of Biology, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
| | - Vitor F O Miranda
- Laboratory of Plant Systematics, Department of Biology, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
| | - Henrik Christensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - John E Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Angelo Berchieri Junior
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal 14884-900, Brazil
| |
Collapse
|
18
|
Yin J, Wang L, Shen R, He J, Li S, Wang H, Cheng Z. The influence of cigR gene on the pathogenicity of Salmonella paratyphi A in vitro and in vivo. FEMS Microbiol Lett 2024; 371:fnae067. [PMID: 39165135 DOI: 10.1093/femsle/fnae067] [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: 06/30/2024] [Revised: 07/26/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024] Open
Abstract
Salmonella Paratyphi A is the causative agent of paratyphoid fever A which is a serious threat to human health in many countries. The cigR gene located in Salmonella pathogenicity island 3 is a type III secretion system 2 effector gene. However, the influence of cigR gene on the pathogenicity of Salmonella Paratyphi A remains unclear. Here, a cigR gene deletion mutant of Salmonella Paratyphi A was constructed and its pathogenic changes were also evaluated. It was found that both the growth and biochemical features have not changed after the loss of cigR, but the absence of cigR significantly enhanced the replication and/or survival ability in phorbol-12-myristate-13-acetate (PMA)-differentiated human macrophage THP-1 cells and in mouse; the proliferative activity and apoptosis of PMA-differentiated THP-1 cell were significantly decreased and increased, respectively, after the lack of cigR gene; and the mutant showed increased virulence to a mouse infection model by decreased half-lethal dose (LD50) value and enhanced the proliferation ratio of bacteria in vivo. These results demonstrated that CigR is an anti-virulence factor and plays an important role in the pathogenicity of Salmonella Paratyphi A.
Collapse
Affiliation(s)
- Junlei Yin
- Medical College, Xinxiang University, Xinxiang, Henan 453003, China
| | - Lijun Wang
- Medical College, Xinxiang University, Xinxiang, Henan 453003, China
| | - Ronghua Shen
- Medical College, Xinxiang University, Xinxiang, Henan 453003, China
| | - Jinjiao He
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453003, China
| | - Shaozu Li
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453003, China
| | - Huajian Wang
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453003, China
| | - Zhao Cheng
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453003, China
| |
Collapse
|
19
|
Meiring JE, Khanam F, Basnyat B, Charles RC, Crump JA, Debellut F, Holt KE, Kariuki S, Mugisha E, Neuzil KM, Parry CM, Pitzer VE, Pollard AJ, Qadri F, Gordon MA. Typhoid fever. Nat Rev Dis Primers 2023; 9:71. [PMID: 38097589 DOI: 10.1038/s41572-023-00480-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 12/18/2023]
Abstract
Typhoid fever is an invasive bacterial disease associated with bloodstream infection that causes a high burden of disease in Africa and Asia. Typhoid primarily affects individuals ranging from infants through to young adults. The causative organism, Salmonella enterica subsp. enterica serovar Typhi is transmitted via the faecal-oral route, crossing the intestinal epithelium and disseminating to systemic and intracellular sites, causing an undifferentiated febrile illness. Blood culture remains the practical reference standard for diagnosis of typhoid fever, where culture testing is available, but novel diagnostic modalities are an important priority under investigation. Since 2017, remarkable progress has been made in defining the global burden of both typhoid fever and antimicrobial resistance; in understanding disease pathogenesis and immunological protection through the use of controlled human infection; and in advancing effective vaccination programmes through strategic multipartner collaboration and targeted clinical trials in multiple high-incidence priority settings. This Primer thus offers a timely update of progress and perspective on future priorities for the global scientific community.
Collapse
Affiliation(s)
- James E Meiring
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
| | - Farhana Khanam
- International Centre for Diarrhoel Disease Research, Dhaka, Bangladesh
| | - Buddha Basnyat
- Oxford University Clinical Research Unit, Kathmandu, Nepal
| | - Richelle C Charles
- Massachusetts General Hospital, Harvard Medical School, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - John A Crump
- Centre for International Health, University of Otago, Dunedin, New Zealand
| | | | - Kathryn E Holt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Emmanuel Mugisha
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher M Parry
- Department of Clinical Sciences and Education, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases and Public Health Modelling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Firdausi Qadri
- International Centre for Diarrhoel Disease Research, Dhaka, Bangladesh
| | - Melita A Gordon
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi.
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
20
|
Vinueza-Burgos C, Medina-Santana J, Maldonado R, Vásquez Y, Lincango L, Villagomez E, Gómez C, Ron-Garrido L, Cevallos-Almeida MB. Evaluation of Virulence of Salmonella Infantis and Salmonella Enteritidis with In Vitro and In Vivo Models. Foodborne Pathog Dis 2023; 20:484-491. [PMID: 37668605 DOI: 10.1089/fpd.2023.0060] [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: 09/06/2023] Open
Abstract
Salmonella Infantis and Enteritidis serovars have been reported as important causes of salmonellosis in humans worldwide. However, the virulence of these two serovars has yet to be compared. To evaluate the virulence of Salmonella Infantis (n = 23) and Salmonella Enteritidis (n = 7), we used two models: the Caco2 cells model (in vitro) and the Galleria mellonella model (in vivo). Additionally, the virulence genes of all tested strains were contrasted with phenotypic outcomes. Results showed that adhesion means were 18.2% for Salmonella Enteritidis and 38.2% for Salmonella Infantis strains. Invasion means were 77.1% for Salmonella Enteritidis and 56.2% for Salmonella Infantis strains. Significant differences were found between serovars in adherence and invasion assays. Mortality rates (58% for Salmonella Enteritidis and 62.6% for Salmonella Infantis) were not significantly different between serotypes. The distribution of virulence genes showed that genes fae (fimbrial adherence determinants) and shdA (nonfimbrial adherence determinants) were only found in Salmonella Infantis strains. On the other hand, the rck gene (invasion) and Plasmid-encoded fimbriae genes (pef A, B, C, D) were present in Salmonella Enteritidis exclusively. In conclusion, this study shows that Salmonella Enteritidis has a higher virulence potential under experimental conditions than Salmonella Infantis. However, more studies are needed to determine the risk that Salmonella Infantis could represent compared with Salmonella Enteritidis. Moreover, other in vivo models should be considered to assess the virulence of these serovars.
Collapse
Affiliation(s)
- Christian Vinueza-Burgos
- Unidad de Investigación en Enfermedades Transmitidas por Alimentos y Resistencia a los Antimicrobianos (UNIETAR), Universidad Central del Ecuador, Quito, Ecuador
| | - Jose Medina-Santana
- Unidad de Investigación en Enfermedades Transmitidas por Alimentos y Resistencia a los Antimicrobianos (UNIETAR), Universidad Central del Ecuador, Quito, Ecuador
| | - Ruben Maldonado
- Laboratorio de Sanidad Animal Agencia de Regulación y Control Fito y Zoosanitario, Quito, Ecuador
| | - Yuly Vásquez
- Laboratorio de Bacteriología y Micología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Central del Ecuador, Quito, Ecuador
| | - Lisseth Lincango
- Laboratorio de Bacteriología y Micología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Central del Ecuador, Quito, Ecuador
| | - Emilia Villagomez
- Laboratorio de Bacteriología y Micología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Central del Ecuador, Quito, Ecuador
| | - Carlos Gómez
- Laboratorio de Bacteriología y Micología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Central del Ecuador, Quito, Ecuador
| | - Lenin Ron-Garrido
- Centro Internacional de Zoonosis, Universidad Central del Ecuador, Quito, Ecuador
| | - María Belén Cevallos-Almeida
- Laboratorio de Bacteriología y Micología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Central del Ecuador, Quito, Ecuador
| |
Collapse
|
21
|
Worley MJ. Salmonella Bloodstream Infections. Trop Med Infect Dis 2023; 8:487. [PMID: 37999606 PMCID: PMC10675298 DOI: 10.3390/tropicalmed8110487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Salmonella is a major foodborne pathogen of both animals and humans. This bacterium is responsible for considerable morbidity and mortality world-wide. Different serovars of this genus cause diseases ranging from self-limiting gastroenteritis to a potentially fatal systemic disease known as enteric fever. Gastrointestinal infections with Salmonella are usually self-limiting and rarely require medical intervention. Bloodstream infections, on the other hand, are often fatal even with hospitalization. This review describes the routes and underlying mechanisms of the extraintestinal dissemination of Salmonella and the chronic infections that sometimes result. It includes information on the pathogenicity islands and individual virulence factors involved in systemic dissemination as well as a discussion of the host factors that mediate susceptibility. Also, the major outbreaks of invasive Salmonella disease in the tropics are described.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
22
|
Abramov VM, Kosarev IV, Machulin AV, Deryusheva EI, Priputnevich TV, Panin AN, Chikileva IO, Abashina TN, Manoyan AM, Ahmetzyanova AA, Ivanova OE, Papazyan TT, Nikonov IN, Suzina NE, Melnikov VG, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN. Ligilactobacillus salivarius 7247 Strain: Probiotic Properties and Anti- Salmonella Effect with Prebiotics. Antibiotics (Basel) 2023; 12:1535. [PMID: 37887236 PMCID: PMC10604316 DOI: 10.3390/antibiotics12101535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
The Ligilactobacillus salivarius 7247 (LS7247) strain, originally isolated from a healthy woman's intestines and reproductive system, has been studied for its probiotic potential, particularly against Salmonella Enteritidis (SE) and Salmonella Typhimurium (ST) as well as its potential use in synbiotics. LS7247 showed high tolerance to gastric and intestinal stress and effectively adhered to human and animal enterocyte monolayers, essential for realizing its probiotic properties. LS7247 showed high anti-Salmonella activity. Additionally, the cell-free culture supernatant (CFS) of LS7247 exhibited anti-Salmonella activity, with a partial reduction upon neutralization with NaOH (p < 0.05), suggesting the presence of anti-Salmonella factors such as lactic acid (LA) and bacteriocins. LS7247 produced a high concentration of LA, reaching 124.0 ± 2.5 mM after 48 h of cultivation. Unique gene clusters in the genome of LS7247 contribute to the production of Enterolysin A and metalloendopeptidase. Notably, LS7247 carries a plasmid with a gene cluster identical to human intestinal strain L. salivarius UCC118, responsible for class IIb bacteriocin synthesis, and a gene cluster identical to porcine strain L. salivarius P1ACE3, responsible for nisin S synthesis. Co-cultivation of LS7247 with SE and ST pathogens reduced their viability by 1.0-1.5 log, attributed to cell wall damage and ATP leakage caused by the CFS. For the first time, the CFS of LS7247 has been shown to inhibit adhesion of SE and ST to human and animal enterocytes (p < 0.01). The combination of Actigen prebiotic and the CFS of LS7247 demonstrated a significant combined effect in inhibiting the adhesion of SE and ST to human and animal enterocytes (p < 0.001). These findings highlight the potential of using the LS7247 as a preventive strategy and employing probiotics and synbiotics to combat the prevalence of salmonellosis in animals and humans caused by multidrug resistant (MDR) strains of SE and ST pathogens.
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; (I.V.K.)
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - 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; (I.V.K.)
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - 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
| | - 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; (I.V.K.)
| | - Irina O. Chikileva
- Laboratory of Cell Immunity, 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; (I.V.K.)
| | - Anna A. Ahmetzyanova
- 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; (I.V.K.)
| | - 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; (I.V.K.)
| | | | - 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
| | - 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
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
23
|
Petano-Duque JM, Rueda-García V, Rondón-Barragán IS. Virulence genes identification in Salmonella enterica isolates from humans, crocodiles, and poultry farms from two regions in Colombia. Vet World 2023; 16:2096-2103. [PMID: 38023281 PMCID: PMC10668553 DOI: 10.14202/vetworld.2023.2096-2103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/11/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Aim Salmonella spp. is frequently found in the digestive tract of birds and reptiles and transmitted to humans through food. Salmonellosis is a public health problem because of pathogenicity variability in strains for virulence factors. This study aimed to identify the virulence genes in Salmonella isolates from humans, crocodiles, broiler cloacas, and broiler carcasses from two departments of Colombia. Materials and Methods This study was conducted on 31 Salmonella enterica strains from humans with gastroenteritis (seven), crocodiles (seven), broiler cloacas (six), and broiler carcasses (12) from Tolima and Santander departments of Colombia, belonging to 21 serotypes. All samples were tested for Salmonella spp. using culture method on selective and non-selective mediums. Extraction of genomic DNA was performed from fresh colonies, DNA quality was verified by spectrophotometry and confirmed by amplification of InvA gene using conventional polymerase chain reaction (PCR). bapA, fimA, icmF, IroB, marT, mgtC, nlpI, oafA, pagN, siiD, spvC, spvR, spvB, Stn, and vexA genes were amplified by PCR. Results The most prevalent gene was bapA (100%), followed by marT (96.77%), mgtC (93.55%), and fimA (83.87%). Likewise, IroB (70.97%), Stn (67.74%), spvR (61.29%), pagN (54.84%), icmF (54.8%), and SiiD (45.16%) were positive for more than 50% of the strains. Furthermore, none of the isolates tested positive for the vexA gene. Salmonella isolates presented 26 virulence profiles. Conclusion This study reported 14 virulence genes in Salmonella spp. isolates from humans with gastroenteritis, crocodiles, and broiler cloacas and carcasses. The distribution of virulence genes differed among sources. This study could help in decision-making by health and sanitary authorities.
Collapse
Affiliation(s)
- Julieth Michel Petano-Duque
- Poultry Research Group, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Santa Helena Highs, Ibagué, Tolima, Colombia
- Research Group in Immunobiology and Pathogenesis, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Santa Helena Highs, Ibagué, Tolima, Colombia
| | - Valentina Rueda-García
- Research Group in Immunobiology and Pathogenesis, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Santa Helena Highs, Ibagué, Tolima, Colombia
| | - Iang Schroniltgen Rondón-Barragán
- Poultry Research Group, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Santa Helena Highs, Ibagué, Tolima, Colombia
- Research Group in Immunobiology and Pathogenesis, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Santa Helena Highs, Ibagué, Tolima, Colombia
| |
Collapse
|
24
|
Bryan-Thomas J, McClear T, Omoregie S. Antimicrobial potential of unstressed and heat stressed Allium sativum. Saudi J Biol Sci 2023; 30:103749. [PMID: 37560481 PMCID: PMC10407899 DOI: 10.1016/j.sjbs.2023.103749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/16/2023] [Accepted: 07/22/2023] [Indexed: 08/11/2023] Open
Abstract
Garlic (Allium sativum) is generally known to be of medicinal value, possessing potentials that include antimicrobial activity, but are often consumed in foods after subjection to cooking heat. The antimicrobial potential of heat stressed garlic may become decreased or lost when cooked, making its medicinal benefit unavailable to consumers. The potential of uncooked and cooked extracts from garlic imported to Jamaica, to inhibit the growth of eight microbes of clinical significance was investigated. Aqueous extracts of fresh garlic of 15 g/100 ml (fw), and dried and pulverized garlic cloves of 12.5 g/100 ml, 25 g/100 ml, 50 g/100 ml, and 100 g/100 ml (dw), were tested for inhibition of microbial growth. Extracts were tested uncooked, and cooked by boiling for 5, 10, and 15 min respectively. Of all the microbes studied, C. albicans incurred the largest zone of inhibition (57.7 ± 0.6 mm at the 100 g/100 ml of the dried extract, F(3, 8) = 51.778, p < 0.001, ω2 = 0.93). Cooking of garlic extracts resulted in statistically significant decreases in zones of inhibition of microbes, as evident in the linear regression and one-way ANOVA analyses, and/or complete loss of microbial inhibition. C. albicans was the most inhibited microbe, followed by E. coli, and Salmonella sp., respectively. The use of uncooked garlic may be the best route for obtaining the greatest antimicrobial potential of garlic against susceptible bacteria and fungi because cooking heat stress resulted in the decrease and complete loss of the antimicrobial potentials of the garlic.
Collapse
Affiliation(s)
- Joneshia Bryan-Thomas
- Department of Medical Technology, Northern Caribbean University, Mandeville, Jamaica
| | - Teena McClear
- Department of Biology, Chemistry and Environmental Science, Northern Caribbean University, Mandeville, Jamaica
| | - Samson Omoregie
- School of Natural and Applied Sciences, University of Technology, Kingston 6, Jamaica
| |
Collapse
|
25
|
Hamblin M, Schade R, Narasimhan R, Monack DM. Salmonella enterica serovar Typhi uses two type 3 secretion systems to replicate in human macrophages and colonize humanized mice. mBio 2023; 14:e0113723. [PMID: 37341487 PMCID: PMC10470537 DOI: 10.1128/mbio.01137-23] [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: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is a human-restricted pathogen that replicates in macrophages. In this study, we investigated the roles of the S. Typhi type 3 secretion systems (T3SSs) encoded on Salmonella pathogenicity islands (SPI)-1 (T3SS-1) and SPI-2 (T3SS-2) during human macrophage infection. We found that mutants of S. Typhi deficient for both T3SSs were defective for intramacrophage replication as measured by flow cytometry, viable bacterial counts, and live time-lapse microscopy. T3SS-secreted proteins PipB2 and SifA contributed to S. Typhi replication and were translocated into the cytosol of human macrophages through both T3SS-1 and T3SS-2, demonstrating functional redundancy for these secretion systems. Importantly, an S. Typhi mutant strain that is deficient for both T3SS-1 and T3SS-2 was severely attenuated in the ability to colonize systemic tissues in a humanized mouse model of typhoid fever. Overall, this study establishes a critical role for S. Typhi T3SSs during its replication within human macrophages and during systemic infection of humanized mice. IMPORTANCE Salmonella enterica serovar Typhi is a human-restricted pathogen that causes typhoid fever. Understanding the key virulence mechanisms that facilitate S. Typhi replication in human phagocytes will enable rational vaccine and antibiotic development to limit the spread of this pathogen. While S. Typhimurium replication in murine models has been studied extensively, there is limited information available about S. Typhi replication in human macrophages, some of which directly conflict with findings from S. Typhimurium murine models. This study establishes that both of S. Typhi's two type 3 secretion systems (T3SS-1 and T3SS-2) contribute to intramacrophage replication and virulence.
Collapse
Affiliation(s)
- Meagan Hamblin
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Ruth Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Ramya Narasimhan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Denise M. Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
26
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. From Eberthella typhi to Salmonella Typhi: The Fascinating Journey of the Virulence and Pathogenicity of Salmonella Typhi. ACS OMEGA 2023; 8:25674-25697. [PMID: 37521659 PMCID: PMC10373206 DOI: 10.1021/acsomega.3c02386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Salmonella Typhi (S. Typhi), the invasive typhoidal serovar of Salmonella enterica that causes typhoid fever in humans, is a severe threat to global health. It is one of the major causes of high morbidity and mortality in developing countries. According to recent WHO estimates, approximately 11-21 million typhoid fever illnesses occur annually worldwide, accounting for 0.12-0.16 million deaths. Salmonella infection can spread to healthy individuals by the consumption of contaminated food and water. Typhoid fever in humans sometimes is accompanied by several other critical extraintestinal complications related to the central nervous system, cardiovascular system, pulmonary system, and hepatobiliary system. Salmonella Pathogenicity Island-1 and Salmonella Pathogenicity Island-2 are the two genomic segments containing genes encoding virulent factors that regulate its invasion and systemic pathogenesis. This Review aims to shed light on a comparative analysis of the virulence and pathogenesis of the typhoidal and nontyphoidal serovars of S. enterica.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Atish Roy Chowdhury
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Debapriya Mukherjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Dipshikha Chakravortty
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
- Centre
for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
27
|
Li Q, Wang L, Xu J, Liu S, Song Z, Chen T, Deng X, Wang J, Lv Q. Quercitrin Is a Novel Inhibitor of Salmonella enterica Serovar Typhimurium Type III Secretion System. Molecules 2023; 28:5455. [PMID: 37513327 PMCID: PMC10383848 DOI: 10.3390/molecules28145455] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The purpose was to screen type III secretory system (T3SS) inhibitors of Salmonella enterica serovar Typhimurium (S. Typhimurium) from natural compounds. The pharmacological activities and action mechanisms of candidate compounds in vivo and in vitro were systematically studied and analyzed. Using a SipA-β-lactamase fusion reporting system, we found that quercitrin significantly blocked the translocation of SipA into eukaryotic host cells without affecting the growth of bacteria. Adhesion and invasion assay showed that quercitrin inhibited S. Typhimurium invasion into host cells and reduced S. Typhimurium mediated host cell damage. β-galactosidase activity detection and Western blot analysis showed that quercitrin significantly inhibited the expression of SPI-1 genes (hilA and sopA) and effectors (SipA and SipC). The results of animal experiments showed that quercitrin significantly reduced colony colonization and alleviated the cecum pathological injury of the infected mice. Small molecule inhibitor quercitrin directly inhibited the function of T3SS and provided a potential antibiotic alternative against S. Typhimurium infection. Importance: T3SS plays a crucial role in the bacterial invasion and pathogenesis of S. Typhimurium. Compared with conventional antibiotics, small molecules could inhibit the virulence factors represented by S. Typhimurium T3SS. They have less pressure on bacterial vitality and a lower probability of producing drug resistance. Our results provide strong evidence for the development of novel inhibitors against S. Typhimurium infection.
Collapse
Affiliation(s)
- Qingjie Li
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinses Medicine, Changchun 130021, China
| | - Lianping Wang
- School of Traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Changchun 132101, China
| | - Jingwen 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, Changchun 130062, China
| | - Shuang Liu
- Jilin Jinziyuan Biotech Inc., Shuangliao 136400, China
| | - Zeyu Song
- 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, Changchun 130062, China
| | - Tingting Chen
- 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, Changchun 130062, China
| | - Xuming Deng
- 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, Changchun 130062, China
| | - Jianfeng 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, Changchun 130062, China
| | - Qianghua Lv
- 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, Changchun 130062, China
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| |
Collapse
|
28
|
Hamblin M, Schade R, Narasimhan R, Monack DM. Salmonella enterica serovar Typhi uses two type 3 secretion systems to replicate in human macrophages and to colonize humanized mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543980. [PMID: 37333307 PMCID: PMC10274799 DOI: 10.1101/2023.06.06.543980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Salmonella enterica serovar Typhi ( S. Typhi) is a human-restricted pathogen that replicates in macrophages. In this study, we investigated the roles of the S. Typhi Type 3 secretion systems (T3SSs) encoded on Salmonella Pathogenicity Islands (SPI) -1 (T3SS-1) and -2 (T3SS-2) during human macrophage infection. We found that mutants of S . Typhi deficient for both T3SSs were defective for intramacrophage replication as measured by flow cytometry, viable bacterial counts, and live time-lapse microscopy. T3SS-secreted proteins PipB2 and SifA contributed to S. Typhi replication and were translocated into the cytosol of human macrophages through both T3SS-1 and -2, demonstrating functional redundancy for these secretion systems. Importantly, an S . Typhi mutant strain that is deficient for both T3SS-1 and -2 was severely attenuated in the ability to colonize systemic tissues in a humanized mouse model of typhoid fever. Overall, this study establishes a critical role for S. Typhi T3SSs during its replication within human macrophages and during systemic infection of humanized mice. Importance Salmonella enterica serovar Typhi is a human-restricted pathogen that causes typhoid fever. Understanding the key virulence mechanisms that facilitate S. Typhi replication in human phagocytes will enable rational vaccine and antibiotic development to limit spread of this pathogen. While S. Typhimurium replication in murine models has been studied extensively, there is limited information available about S. Typhi replication in human macrophages, some of which directly conflicts with findings from S. Typhimurium murine models. This study establishes that both of S. Typhi's two Type 3 Secretion Systems (T3SS-1 and -2) contribute to intramacrophage replication and virulence.
Collapse
|
29
|
Yue M, Zhao X, Hu X, Liu D, Li X, Wu Y. Genomic characterization of a multidrug-resistant Salmonella Rissen ST469 carrying bla CTX-M-55 gene and Tn6777 isolated from a paediatric patient in China. J Glob Antimicrob Resist 2023; 33:238-241. [PMID: 36878464 DOI: 10.1016/j.jgar.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/07/2023] Open
Abstract
OBJECTIVES Here we report a complete genome sequence of a multidrug-resistant Salmonella Rissen, carrying blaCTX-M-55 and Tn6777, isolated from a Chinese paediatric patient. METHODS Whole genome of S. Rissen S1905 was sequenced using the Oxford Nanopore MinION and Illumina NovaSeq 6000 platforms. Unicycler was used to perform a de novo assembly of Illumina and Nanopore reads. The genome sequence was annotated using the NCBI Prokaryotic Genome Annotation Pipeline. In silico multilocus sequence typing, plasmid replicons, antimicrobial resistance genes and virulence factors were identified from the genome sequence by multiple bioinformatics tools. Core genome multilocus sequence typing analysis between S. Rissen S1905 and all retrieved from the NCBI GenBank database was performed using BacWGSTdb 2.0 server. RESULTS Six contigs totaling 5 056 896 bp make up the complete genome sequence of S. Rissen S1905, which includes 1 chromosome and 5 plasmids. The blaCTX-M-55 was embedded in the ISEcp1-blaCTX-M-55-wbuC transposition unit located in an 85 991-bp IncI1 plasmid. However, the pco-sil operon and other eight antimicrobial resistance genes were carried by Tn6777 in the chromosome. There are 162 virulence genes in S1905. S. Rissen S1905 belongs to ST469; the closest relative was another isolate originating from a human faecal specimen in Shanghai, China, which differed by 60 core genome multilocus sequence type alleles. CONCLUSION These data on the multidrug-resistant S. Rissen carrying blaCTX-M-55 and Tn6777 can provide a foundation for further studies on the molecular epidemiological characteristics, pathogenicity, antimicrobial resistance mechanisms, and dissemination mechanism of Salmonella.
Collapse
Affiliation(s)
- Meina Yue
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Xinfeng Zhao
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Xue Hu
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Di Liu
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Xiaoyu Li
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China
| | - Yidong Wu
- Department of Clinical Laboratory, Hangzhou Children's Hospital, Hangzhou, Zhejiang, China; Department of Clinical Laboratory, Hangzhou Ninth People's Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
30
|
Zhang W, Liang G, Cheng Z, Guo Y, Jiang B, Liu T, Liao W, Lu Q, Wen G, Zhang T, Luo Q. Flos populi (Male Inflorescence of Populus tomentosa Carrière) Aqueous Extract Suppresses Salmonella Pullorum Infection by Affecting T3SS-1. Pathogens 2023; 12:790. [PMID: 37375480 DOI: 10.3390/pathogens12060790] [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: 03/09/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Pullorum disease, caused by Salmonella Pullorum (S. Pullorum), is one of the most serious infectious diseases in the poultry industry. Flos populi is traditionally used in Eastern Asian countries to treat various intestinal diseases. However, the anti-infection mechanism of Flos populi is not very clear. In this study, we evaluated the anti-infective effects on S. Pullorum of Flos populi aqueous extract (FPAE) in chickens. FPAE significantly reduced S. Pullorum growth in vitro. At the cellular level, FPAE reduced S. Pullorum adhesion and invasion on DF-1 cells but did not affect its intracellular survival or replication in macrophages. Further investigation revealed that FPAE inhibited the transcription of T3SS-1 genes, which is the main virulence factor that mediates S. Pullorum adhesion and invasion in host cells. The results suggest that the anti-infective effect of FPAE likely occurs through the inhibition of S. Pullorum T3SS-1, thereby impairing its ability to adhere to and invade cells. Further, we evaluated its therapeutic effect on animal models (Jianghan domestic chickens) and found that FPAE reduced the bacterial loads in organs and decreased the mortality and weight loss of infected chickens. Our findings provide novel insights into the potential development of FPAE against S. Pullorum as an effective anti-virulence therapeutic substitute for antibiotics.
Collapse
Affiliation(s)
- Wenting Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Guixing Liang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Yunqing Guo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Boda Jiang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Tingjiang Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Weidong Liao
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qin Lu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Guoyuan Wen
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Tengfei Zhang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qingping Luo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
- Hubei Hongshan Laboratory, Wuhan 430064, China
| |
Collapse
|
31
|
Gibbs KD, Wang L, Yang Z, Anderson CE, Bourgeois JS, Cao Y, Gaggioli MR, Biel M, Puertollano R, Chen CC, Ko DC. Human variation impacting MCOLN2 restricts Salmonella Typhi replication by magnesium deprivation. CELL GENOMICS 2023; 3:100290. [PMID: 37228749 PMCID: PMC10203047 DOI: 10.1016/j.xgen.2023.100290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/24/2023] [Accepted: 02/27/2023] [Indexed: 05/27/2023]
Abstract
Human genetic diversity can reveal critical factors in host-pathogen interactions. This is especially useful for human-restricted pathogens like Salmonella enterica serovar Typhi (S. Typhi), the cause of typhoid fever. One key defense during bacterial infection is nutritional immunity: host cells attempt to restrict bacterial replication by denying bacteria access to key nutrients or supplying toxic metabolites. Here, a cellular genome-wide association study of intracellular replication by S. Typhi in nearly a thousand cell lines from around the world-and extensive follow-up using intracellular S. Typhi transcriptomics and manipulation of magnesium availability-demonstrates that the divalent cation channel mucolipin-2 (MCOLN2 or TRPML2) restricts S. Typhi intracellular replication through magnesium deprivation. Mg2+ currents, conducted through MCOLN2 and out of endolysosomes, were measured directly using patch-clamping of the endolysosomal membrane. Our results reveal Mg2+ limitation as a key component of nutritional immunity against S. Typhi and as a source of variable host resistance.
Collapse
Affiliation(s)
- Kyle D. Gibbs
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Zhuo Yang
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline E. Anderson
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jeffrey S. Bourgeois
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Yanlu Cao
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Margaret R. Gaggioli
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, & Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
32
|
Teklemariam AD, Al-Hindi RR, Albiheyri RS, Alharbi MG, Alghamdi MA, Filimban AAR, Al Mutiri AS, Al-Alyani AM, Alseghayer MS, Almaneea AM, Albar AH, Khormi MA, Bhunia AK. Human Salmonellosis: A Continuous Global Threat in the Farm-to-Fork Food Safety Continuum. Foods 2023; 12:foods12091756. [PMID: 37174295 PMCID: PMC10178548 DOI: 10.3390/foods12091756] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Salmonella is one of the most common zoonotic foodborne pathogens and a worldwide public health threat. Salmonella enterica is the most pathogenic among Salmonella species, comprising over 2500 serovars. It causes typhoid fever and gastroenteritis, and the serovars responsible for the later disease are known as non-typhoidal Salmonella (NTS). Salmonella transmission to humans happens along the farm-to-fork continuum via contaminated animal- and plant-derived foods, including poultry, eggs, fish, pork, beef, vegetables, fruits, nuts, and flour. Several virulence factors have been recognized to play a vital role in attaching, invading, and evading the host defense system. These factors include capsule, adhesion proteins, flagella, plasmids, and type III secretion systems that are encoded on the Salmonella pathogenicity islands. The increased global prevalence of NTS serovars in recent years indicates that the control approaches centered on alleviating the food animals' contamination along the food chain have been unsuccessful. Moreover, the emergence of antibiotic-resistant Salmonella variants suggests a potential food safety crisis. This review summarizes the current state of the knowledge on the nomenclature, microbiological features, virulence factors, and the mechanism of antimicrobial resistance of Salmonella. Furthermore, it provides insights into the pathogenesis and epidemiology of Salmonella infections. The recent outbreaks of salmonellosis reported in different clinical settings and geographical regions, including Africa, the Middle East and North Africa, Latin America, Europe, and the USA in the farm-to-fork continuum, are also highlighted.
Collapse
Affiliation(s)
- Addisu D Teklemariam
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rashad R Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed S Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mona G Alharbi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashail A Alghamdi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani A R Filimban
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdullah S Al Mutiri
- Laboratory Department, Saudi Food and Drug Authority, Riyadh 12843, Saudi Arabia
| | - Abdullah M Al-Alyani
- Laboratory Department, Saudi Food and Drug Authority, Jeddah 22311, Saudi Arabia
| | - Mazen S Alseghayer
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulaziz M Almaneea
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Monitoring and Risk Assessment Department, Saudi Food and Drug Authority, Riyadh 13513, Saudi Arabia
| | - Abdulgader H Albar
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Microbiology and Medical Parasitology, Faculty of Medicine, Jeddah University, Jeddah 23218, Saudi Arabia
| | - Mohsen A Khormi
- Department of Biological Sciences, Faculty of Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Interdisciplinary Life Science Program (PULSe), West Lafayette, IN 47907, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
33
|
Wang BX, Butler DS, Hamblin M, Monack DM. One species, different diseases: the unique molecular mechanisms that underlie the pathogenesis of typhoidal Salmonella infections. Curr Opin Microbiol 2023; 72:102262. [PMID: 36640585 PMCID: PMC10023398 DOI: 10.1016/j.mib.2022.102262] [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: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023]
Abstract
Salmonella enterica is one of the most widespread bacterial pathogens found worldwide, resulting in approximately 100 million infections and over 200 000 deaths per year. Salmonella isolates, termed 'serovars', can largely be classified as either nontyphoidal or typhoidal Salmonella, which differ in regard to disease manifestation and host tropism. Nontyphoidal Salmonella causes gastroenteritis in many hosts, while typhoidal Salmonella is human-restricted and causes typhoid fever, a systemic disease with a mortality rate of up to 30% without treatment. There has been considerable interest in understanding how different Salmonella serovars cause different diseases, but the molecular details that underlie these infections have not yet been fully characterized, especially in the case of typhoidal Salmonella. In this review, we highlight the current state of research into understanding the pathogenesis of both nontyphoidal and typhoidal Salmonella, with a specific interest in serovar-specific traits that allow human-adapted strains of Salmonella to cause enteric fever. Overall, a more detailed molecular understanding of how different Salmonella isolates infect humans will provide critical insights into how we can eradicate these dangerous enteric pathogens.
Collapse
Affiliation(s)
- Benjamin X Wang
- Department of Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Daniel Sc Butler
- Department of Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Meagan Hamblin
- Department of Microbiology & Immunology, Stanford University, Stanford, CA, USA
| | - Denise M Monack
- Department of Microbiology & Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Kambouris ME. Global Catastrophic Biological Risks in the Post-COVID-19 World: Time to Act Is Now. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:153-170. [PMID: 36946656 DOI: 10.1089/omi.2022.0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Global Catastrophic Biological Risks (GCBRs) refer to events with biological agents that can result in unprecedented or catastrophic disasters that are beyond the collective response-abilities of nation-states and the existing governance instruments of global governance and international affairs. This article offers a narrative review, with a view to new hypothesis development to rethink GCBRs after coronavirus disease 2019 (COVID-19) so as to better prepare for future pandemics and ecological crises, if not to completely prevent them. To determine GCBRs' spatiotemporal contexts, define causality, impacts, differentiate the risk and the event, would improve theorization of GCBRs compared to the impact-centric current definition. This could in turn lead to improvements in preparedness, response, allocation of resources, and possibly deterrence, while actively discouraging lack of due biosecurity diligence. Critical governance of GCBRs in ways that unpack the political power-related dimensions could be particularly valuable because the future global catastrophic events might be different in quality, scale, and actors. Theorization of GCBRs remains an important task going forward in the 21st century in ways that draw from experiences in the field, while integrating flexibility, versatility, and critically informed responses to GCBRs.
Collapse
|
35
|
Sharma S, Solanki V, Tiwari V. Reverse vaccinology approach to design a vaccine targeting membrane lipoproteins of Salmonella typhi. J Biomol Struct Dyn 2023; 41:954-969. [PMID: 34939517 DOI: 10.1080/07391102.2021.2015443] [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] [Indexed: 01/11/2023]
Abstract
Typhoid fever caused by Salmonella is one of the major health issues worldwide, resulting in millions of cases and has very high rates of morbidities. The therapeutic approaches need to be updated for the effective elimination of the bacterial pathogen. The designing of the multiepitope vaccine against Salmonella using comparative proteomics and reverse vaccinology has covered up all the epitopes that induce sufficient immune responses in the host body. Out of the 4293 proteins, 15 outer membrane proteins have been selected based on their antigenicity, low transmembrane helix (<1), and virulence-associated factors. With the help of the reverse vaccinology approach, the epitopes of MHC Class I, Class II, and B-cell with antigenic, low toxicity, and that have the potential to generate immunogenic response have been identified. Based on the comparative analysis of all the epitopes, a multiepitope-based construct has been designed. Based on physicochemical properties and docking scores for HLA and TLR4, the VC5 construct has been selected, and the molecular dynamic simulation studies have confirmed their interaction. The dissociation constant of the VC5 and TLR4 was found to be 3.1 x 10-9. Different immune cell activation has been analyzed, representing the potentiality of the VC5 construct as an effective vaccine target. In silico cloning of VC5 in pET28a has also been performed, which requires experimental validation. Therefore, the present study designs a multi-epitope vaccine VC5 targeted to the membrane lipoproteins of Salmonella typhi.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saroj Sharma
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Vandana Solanki
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
36
|
Resistance Mechanism and Physiological Effects of Microcin Y in Salmonella enterica subsp. enterica Serovar Typhimurium. Microbiol Spectr 2022; 10:e0185922. [PMID: 36453909 PMCID: PMC9769762 DOI: 10.1128/spectrum.01859-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Salmonella bacteria pose a significant threat to animal husbandry and human health due to their virulence and multidrug resistance. The lasso peptide MccY is a recently discovered antimicrobial peptide that acts against various serotypes of Salmonella. In this study, we further explore the resistance mechanism and activity of MccY. Mutants of Ton system genes, including tonB, exbB, and exbD, in Salmonella enterica subsp. enterica serovar Typhimurium were constructed, and the MICs to MccY exhibited significant increases in these deletion mutants compared to the MIC of the parent strain. Subsequently, MccY resistance was quantitatively analyzed, and these mutants also showed greatly reduced rates of killing, even with a high concentration of MccY. In addition, a minimal medium with low iron environment enhanced the sensitivity of these mutants to MccY. Measurements of a series of physiological indicators, including iron utilization, biofilm formation, and motility, demonstrated that MccY may decrease the virulence of S. Typhimurium. Transcriptomic analysis showed that iron utilization, biofilm formation, flagellar assembly, and virulence-related genes were downregulated to varying degrees when S. Typhimurium was treated with MccY. In conclusion, deletion of Ton system genes resulted in resistance to MccY and the susceptibility of these mutants to MccY was increased and differed under a low-iron condition. This lasso peptide can alter multiple physiological properties of S. Typhimurium. Our study will contribute to improve the knowledge and understanding of the mechanism of MccY resistance in Salmonella strains. IMPORTANCE The resistance of Salmonella to traditional antibiotics remains a serious challenge. Novel anti-Salmonella drugs are urgently needed to address the looming crisis. The newly identified antimicrobial peptide MccY shows broad prospects for development and application because of its obvious antagonistic effect on various serotypes of Salmonella. However, our previous study showed that the peptide could confer resistance to Salmonella by disrupting the receptor gene fhuA. In this study, we further explored the potential resistance mechanism of MccY and demonstrated the importance of the Salmonella Ton complex for MccY transport. Disruption in Ton system genes resulted in S. Typhimurium resistance to this peptide, and MccY could alter multiple bacterial physiological properties. In summary, this study further explored the resistance mechanism and antibacterial effect of MccY in S. Typhimurium and provided a scientific basis for its development and application.
Collapse
|
37
|
Kanaan MHG, Khalil ZK, Khashan HT, Ghasemian A. Occurrence of virulence factors and carbapenemase genes in Salmonella enterica serovar Enteritidis isolated from chicken meat and egg samples in Iraq. BMC Microbiol 2022; 22:279. [PMID: 36418940 PMCID: PMC9682753 DOI: 10.1186/s12866-022-02696-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Food-borne infections mainly due to Salmonella enterica serovar Enteritidis (S. Enteritidis) are major concerns worldwide. S. Enteritidis isolates may serve as reservoirs for spreading antimicrobial drug resistance genes including carbapenemases. This study aimed to screen the occurrence of virulence factors, carbapenemases, and antibiotic resistance genes in S. Enteritidis isolated from chicken meat and eggs in Iraq. RESULTS In total, 1000 non-duplicated chicken meat and 1000 egg samples were collected during 2019-2020. Presumptive S. Enteritidis isolates were initially identified by standard bacteriology tests and then were confirmed using polymerase chain reaction (PCR). Carbapenem resistance was detected using the disk diffusion method. Virulence and carbapenemase genes were screened using the PCR method. In total, 100 (5.0%) S. Enteritidis isolates were identified from 2000 samples collected using phenotypic and molecular methods. These isolates were identified from 4.9% chicken meat (n = 49/1000) and 5.1% egg (n = 51/1000) samples, respectively. The most and the least susceptibility was found to gentamicin and ceftazidime antibiotics, respectively. The prevalence of different virulence factors were as follows: phoP/Q (40.0%), traT (30.0%), stn (22.0%), slyA (11.0%), and sopB (9.0%). Among 20 carbapenem-resistant S. Enteritidis isolates, the most predominant carbapenemase gene was blaIMP (35.0%, n = 7), followed by blaOXA-48-like (25.0%, n = 5), and blaNDM (10.0%, n = 2), while the blaKPC and blaVIM genes were not detected. The coexistence of blaIMP, blaOXA-48-like, and blaNDM genes was determined in two isolates. The prevalence of different antibiotic resistance genes were as follows: tetA (87.1%), tetB (87.1%), dfrA1 (77.6%), and sul1 (83.6%). CONCLUSION Considering the existence of carbapenem-resistant S. Enteritidis harboring different virulence and antibiotic resistance genes in chicken meat and egg samples, adherence to proper hygienic conditions should be considered.
Collapse
Affiliation(s)
- Manal Hadi Ghaffoori Kanaan
- grid.510261.10000 0004 7474 9372Department of Agriculture, Technical Institute of Suwaria, Middle Technical University, Baghdad, Iraq
| | - Zena Kassem Khalil
- grid.510261.10000 0004 7474 9372Optometry Department, Medical Technical Institute Al-Mansor, Middle Technical University, Baghdad, Iraq
| | - Hawazin Thamir Khashan
- grid.411498.10000 0001 2108 8169Department of Veterinary Public Health, Food Hygiene, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Abdolmajid Ghasemian
- grid.411135.30000 0004 0415 3047Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
38
|
Zhang X, Liu B, Ding X, Bin P, Yang Y, Zhu G. Regulatory Mechanisms between Quorum Sensing and Virulence in Salmonella. Microorganisms 2022; 10:2211. [PMID: 36363803 PMCID: PMC9693372 DOI: 10.3390/microorganisms10112211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 08/28/2023] Open
Abstract
Salmonella is a foodborne pathogen that causes enterogastritis among humans, livestock and poultry, and it not only causes huge economic losses for the feed industry but also endangers public health around the world. However, the prevention and treatment of Salmonella infection has remained poorly developed because of its antibiotic resistance. Bacterial quorum sensing (QS) system is an intercellular cell-cell communication mechanism involving multiple cellular processes, especially bacterial virulence, such as biofilm formation, motility, adherence, and invasion. Therefore, blocking the QS system may be a new strategy for Salmonella infection independent of antibiotic treatment. Here, we have reviewed the central role of the QS system in virulence regulation of Salmonella and summarized the most recent advances about quorum quenching (QQ) in virulence attenuation during Salmonella infection. Unraveling the complex relationship between QS and bacterial virulence may provide new insight into the therapy of pathogen infection.
Collapse
Affiliation(s)
- Xiaojie Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Baobao Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xueyan Ding
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Peng Bin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yang Yang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
39
|
Li Y, Teng L, Xu X, Li X, Peng X, Zhou X, Du J, Tang Y, Jiang Z, Wang Z, Jia C, Müller A, Kehrenberg C, Wang H, Wu B, Weill F, Yue M. A nontyphoidal Salmonella serovar domestication accompanying enhanced niche adaptation. EMBO Mol Med 2022; 14:e16366. [PMID: 36172999 PMCID: PMC9641423 DOI: 10.15252/emmm.202216366] [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: 05/24/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Invasive nontyphoidal Salmonella (iNTS) causes extraintestinal infections with ~15% case fatality in many countries. However, the mechanism by which iNTS emerged in China remains unaddressed. We conducted clinical investigations of iNTS infection with recurrent treatment failure, caused by underreported Salmonella enterica serovar Livingstone (SL). Genomic epidemiology demonstrated five clades in the SL population and suggested that the international animal feed trade was a likely vehicle for their introduction into China, as evidenced by multiple independent transmission incidents. Importantly, isolates from Clade-5-I-a/b, predominant in China, showed an invasive nature in mice, chicken and zebrafish infection models. The antimicrobial susceptibility testing revealed most isolates (> 96%) in China are multidrug-resistant (MDR). Overall, we offer exploiting genomics in uncovering international transmission led by the animal feed trade and highlight an emerging hypervirulent clade with increased resistance to frontline antibiotics.
Collapse
Affiliation(s)
- Yan Li
- Hainan Institute of Zhejiang UniversitySanyaChina
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
- Zhejiang Provincial Key Laboratory of Preventive Veterinary MedicineHangzhouChina
| | - Lin Teng
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Xuebin Xu
- Shanghai Municipal Center for Disease Control and PreventionShanghaiChina
| | - Xiaomeng Li
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Xianqi Peng
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Xiao Zhou
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Jiaxin Du
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Yanting Tang
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Zhijie Jiang
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Zining Wang
- Hainan Institute of Zhejiang UniversitySanyaChina
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Chenghao Jia
- Hainan Institute of Zhejiang UniversitySanyaChina
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
| | - Anja Müller
- Institute for Veterinary Food Science, Faculty of Veterinary MedicineJustus‐Liebig University GiessenGiessenGermany
| | - Corinna Kehrenberg
- Institute for Veterinary Food Science, Faculty of Veterinary MedicineJustus‐Liebig University GiessenGiessenGermany
| | - Haoqiu Wang
- Hangzhou Center for Disease Control and PreventionHangzhouChina
| | - Beibei Wu
- Zhejiang Province Center for Disease Control and PreventionHangzhouChina
| | | | - Min Yue
- Hainan Institute of Zhejiang UniversitySanyaChina
- Institute of Preventive Veterinary Science & Department of Veterinary MedicineZhejiang University College of Animal SciencesHangzhouChina
- Zhejiang Provincial Key Laboratory of Preventive Veterinary MedicineHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
40
|
Abstract
Rapid qualitative and quantitative detection of Salmonella typhimurium (S. typhimurium) takes an important role in ensuring food safety. Herein, a colorimetric assay aptasensor for S. typhimurium utilizing intrinsic peroxidase-like activity of gold nanoparticles embedded spherical covalent organic framework and the affinity and specificity of S. typhimurium-aptamer has been explored. This aptasensor can capture the S. typhimurium via the selective binding effect of aptamer, and the catalytically active sites were shielded. As a result, the colorimetric signals of the 3,3′,5,5′-tetramethylbenzidine-H2O2 system were turned off. Under optimum conditions, the aptasensor gave a linear response over the range of 10 to 107 CFU/mL for S. typhimurium. The detection limit of 7 CFU/mL was obtained within 45 min and was effectively applied to detect S. typhimurium in milk and lake water samples with recoveries in the range from 96.4 to 101.0%. More importantly, combined with a self-developed smartphone-based image analysis system, the proposed aptasensor can be used for point-of-care testing applications.
Collapse
|
41
|
Neupane DP, Ahn C, Yang YA, Lee GY, Song J. Malnutrition and maternal vaccination against typhoid toxin. PLoS Pathog 2022; 18:e1010731. [PMID: 35960787 PMCID: PMC9401117 DOI: 10.1371/journal.ppat.1010731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/24/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
Children are particularly susceptible to typhoid fever caused by the bacterial pathogen Salmonella Typhi. Typhoid fever is prevalent in developing countries where diets can be less well-balanced. Here, using a murine model, we investigated the role of the macronutrient composition of the diet in maternal vaccination efficacies of two subunit vaccines targeting typhoid toxin: ToxoidVac and PltBVac. We found that maternal vaccinations protected all offspring against a lethal-dose typhoid toxin challenge in a balanced, normal diet (ND) condition, but the declined protection in a malnourished diet (MD) condition was observed in the PltBVac group. Despite the comparable antibody titers in both MD and ND mothers, MD offspring had a significantly lower level of typhoid toxin neutralizing antibodies than their ND counterparts. We observed a lower expression of the neonatal Fc receptor on the yolk sac of MD mothers than in ND mothers, agreeing with the observed lower antibody titers in MD offspring. Protein supplementation to MD diets, but not fat supplementation, increased FcRn expression and protected all MD offspring from the toxin challenge. Similarly, providing additional typhoid toxin-neutralizing antibodies to MD offspring was sufficient to protect all MD offspring from the toxin challenge. These results emphasize the significance of balanced/normal diets for a more effective maternal vaccination transfer to their offspring.
Collapse
Affiliation(s)
- Durga P. Neupane
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Changhwan Ahn
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Yi-An Yang
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Gi Young Lee
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Jeongmin Song
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
42
|
Liu Z, Wang L, Yu Y, Fotin A, Wang Q, Gao P, Zhang Y, Fotina T, Ma J. SteE Enhances the Virulence of Salmonella Pullorum in Chickens by Regulating the Inflammation Response. Front Vet Sci 2022; 9:926505. [PMID: 35909683 PMCID: PMC9330158 DOI: 10.3389/fvets.2022.926505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/16/2022] [Indexed: 11/18/2022] Open
Abstract
Salmonella enterica serovar Pullorum (S. Pullorum) is a host-specific pathogen, which causes acute gastroenteritis with high mortality in poultry. However, the association between steE, encoded by type III secretion system 2, and Salmonella virulence is not well-understood. To elucidate the functions of steE in S. Pullorum, ΔsteE strain was constructed using the λ-Red recombination technology. Compared to that in the wild-type, the deletion of steE in S. Pullorum reduced bacterial invasion, proliferation, and late apoptosis in the infected HD-11 cells. In addition, we analyzed the mRNA expression levels of effector genes and cytokines by qRT-PCR. SteE was associated with the regulation of various effector genes and inflammatory cytokines in HD-11 cells during S. Pullorum infection. The wild-type effector steE promoted the expression of anti-inflammatory cytokines (IL-4 and IL-10) and reduced that of pro-inflammatory cytokines (IL-1β, IL-6, and IL-12) compared to that in the ΔsteE-infected HD-11 cells and chicken spleens. Results from the chicken infection model showed that the deletion of steE resulted in significantly decreased colonization and long-term survival of the bacteria and alleviated pathological lesions compared to those in the wild-type. Further, steE increased the virulence of S. Pullorum in chickens by regulating the expression of inflammatory cytokines. Our findings provide insights into the persistent infection and autoimmunity associated with steE in S. Pullorum.
Collapse
Affiliation(s)
- Zhike Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Li Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yan Yu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Anatoliy Fotin
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Qiuxia Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Pei Gao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yanhong Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Tetiana Fotina
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
- *Correspondence: Tetiana Fotina
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Jinyou Ma
| |
Collapse
|
43
|
Zhang J, Liu J, Chen C, Wang Y, Chen X, Li X, Xu F. Resistance and Pathogenicity of Salmonella Thompson Isolated from Incubation End of a Poultry Farm. Vet Sci 2022; 9:vetsci9070349. [PMID: 35878365 PMCID: PMC9323645 DOI: 10.3390/vetsci9070349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/25/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Non-typhoid Salmonella is the general term of Salmonella other than typhoid and paratyphoid, which often causes foodborne gastroenteritis in humans, but some serotypes have been proved to be pathogenic to poultry. Salmonella Enterica and Salmonella Typhimurium are the common serotypes pathogenic to poultry and have been systematically studied, but other serotypes have rarely been studied. During Salmonella surveillance in farms, we discovered by chance that Salmonella Thompson, a common non-typhoid Salmonella, is also pathogenic to avian embryos. Therefore, this study aimed to explore antimicrobial resistance and pathogenicity of clinical S. Thompson. Firstly, we found that the core-genome multilocus sequence typing of 14 clinical S. Thompson was consistent with two strains of S. Thompson from humans in China. Secondly, the antimicrobial resistance gene analysis demonstrated that all strains carried the polymyxin resistance gene mcr-9, which had not appeared resistance phenotype. Meanwhile, many essential virulence genes were also found in each S. Thompson isolate. Finally, the bacterial inoculation experiment revealed that clinical S. Thompson was highly pathogenic to newborn chicks after yolk sac inoculation. This study suggests that Salmonella Thompson can circulate between humans and poultry farms and transmit drug resistance genes and demonstrated that Salmonella Thompson is highly pathogenic to chicks and should be guarded against in the hatching stage of poultry farms. Abstract Salmonella Thompson, an important foodborne pathogen, is rarely found to be pathogenic to poultry. Accidentally, S. Thompson was found to be pathogenic to embryos of white feather broiler at a poultry farm in China. Therefore, this study aimed to explore antimicrobial resistance and pathogenicity of clinical S. Thompson isolated from dead poultry embryos. The phylogenetic tree based on 16S rRNA and seven housekeeping genes showed that the 14 clinical S. Thompson were closely related. The core-genome multilocus sequence typing of 14 clinical S. Thompson based on whole-genome sequencing was cgST-12774, consistent with the only two strains of S. Thompson from humans in China as reported in the NCBI database. The antimicrobial resistance gene analysis demonstrated that all strains carried aac(6′)-Iaa and the polymyxin resistance gene mcr-9. Antimicrobial sensitivity tests for 18 antibiotics showed that S. Thompson isolates displayed resistance against streptomycin (100%), ampicillin (35.7%), and doxycycline (14.3%), but sensitivity to polymyxin B, proving that the mcr-9 gene had not appeared resistance phenotype. Virulence genes Salmonella pathogenicity island (SPI) SPI1-5, type I fimbriae gene (fimA), flagellar assembly genes (bcfC, flhD, fliA, fliC, fljB, flgK, and lpfC), and other virulence genes (iroN, pagC, and cigR) were found in each S. Thompson isolate. Additionally, the bacterial inoculation experiment with 1-day-old chicks revealed that clinical S. Thompson was highly pathogenic to newborn chicks after yolk sac inoculation. This study highlighted that the S. Thompson isolated from poultry embryos and the S. Thompson causing human foodborne diarrhea in some parts of China belong to the same cgMLST typology (cgST-12774) and showed the pathogenicity of this clinical S. Thompson to chicks.
Collapse
|
44
|
Abstract
Pyroptosis, a regulated form of pro-inflammatory cell death, is characterised by cell lysis and by the release of cytokines, damage- and pathogen-associated molecular patterns. It plays an important role during bacterial infection, where it can promote an inflammatory response and eliminate the replicative niche of intracellular pathogens. Recent work, using a variety of bacterial pathogens, has illuminated the versatility of pyroptosis, revealing unexpected and important concepts underlying host defence. In this Review, we overview the molecular mechanisms underlying pyroptosis and discuss their role in host defence, from the single cell to the whole organism. We focus on recent studies using three cellular microbiology paradigms - Mycobacterium tuberculosis, Salmonella Typhimurium and Shigella flexneri - that have transformed the field of pyroptosis. We compare insights discovered in tissue culture, zebrafish and mouse models, highlighting the advantages and disadvantages of using these complementary infection models to investigate pyroptosis and for modelling human infection. Moving forward, we propose that in-depth knowledge of pyroptosis obtained from complementary infection models can better inform future studies using higher vertebrates, including humans, and help develop innovative host-directed therapies to combat bacterial infection.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
45
|
Sanchez-Garrido J, Ruano-Gallego D, Choudhary JS, Frankel G. The type III secretion system effector network hypothesis. Trends Microbiol 2022; 30:524-533. [PMID: 34840074 DOI: 10.1016/j.tim.2021.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/18/2022]
Abstract
Type III secretion system (T3SS) effectors are key virulence factors that underpin the infection strategy of many clinically important Gram-negative pathogens, including Salmonella enterica, Shigella spp., enteropathogenic and enterohemorrhagic Escherichia coli and their murine equivalent, Citrobacter rodentium. The cellular processes or proteins targeted by the effectors can be common to multiple pathogens or pathogen-specific. The main approach to understanding T3SS-mediated pathogenesis has been to determine the contribution of one effector at a time, with the aim of piecing together individual functions and unveiling infection mechanisms. However, in contrast to this prevailing approach, simultaneous deletion of multiple effectors revealed that they function as an interconnected network in vivo, uncovering effector codependency and context-dependent effector essentiality. This paradigm shift in T3SS biology is at the heart of this opinion article.
Collapse
Affiliation(s)
- Julia Sanchez-Garrido
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK.
| | - David Ruano-Gallego
- Department of Molecular Evolution, Centro de Astrobiología, Instituto Nacional de Técnica Aeroespacial-Consejo Superior de Investigaciones Científicas (INTA-CSIC), Madrid, Spain.
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, UK
| | - Gad Frankel
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| |
Collapse
|
46
|
Maes M, Sikorski MJ, Carey ME, Higginson EE, Dyson ZA, Fernandez A, Araya P, Tennant SM, Baker S, Lagos R, Hormazábal JC, Levine MM, Dougan G. Whole genome sequence analysis of Salmonella Typhi provides evidence of phylogenetic linkage between cases of typhoid fever in Santiago, Chile in the 1980s and 2010-2016. PLoS Negl Trop Dis 2022; 16:e0010178. [PMID: 35767580 PMCID: PMC9275700 DOI: 10.1371/journal.pntd.0010178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/12/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022] Open
Abstract
Typhoid fever epidemiology was investigated rigorously in Santiago, Chile during the 1980s, when Salmonella enterica serovar Typhi (S. Typhi) caused seasonal, hyperendemic disease. Targeted interventions reduced the annual typhoid incidence rates from 128–220 cases/105 population occurring between 1977–1984 to <8 cases/105 from 1992 onwards. As such, Santiago represents a contemporary example of the epidemiologic transition of an industrialized city from amplified hyperendemic typhoid fever to a period when typhoid is no longer endemic. We used whole genome sequencing (WGS) and phylogenetic analysis to compare the genotypes of S. Typhi cultured from acute cases of typhoid fever occurring in Santiago during the hyperendemic period of the 1980s (n = 74) versus the nonendemic 2010s (n = 80) when typhoid fever was rare. The genotype distribution between “historical” (1980s) isolates and “modern” (2011–2016) isolates was similar, with genotypes 3.5 and 2 comprising the majority of isolations, and 73/80 (91.3%) of modern isolates matching a genotype detected in the 1980s. Additionally, phylogenomically ‘ancient’ genotypes 1.1 and 1.2.1, uncommon in the global collections, were also detected in both eras, with a notable rise amongst the modern isolates. Thus, genotypes of S. Typhi causing acute illness in the modern nonendemic era match the genotypes circulating during the hyperendemic 1980s. The persistence of historical genotypes may be explained by chronic typhoid carriers originally infected during or before the 1980s. Studies of Salmonella Typhi (the cause of typhoid fever) rarely include isolates collected both before and after the interruption of hyperendemic transmission because this typically occurred decades before modern bacteria preservation methods. After substantial reduction in disease, it was assumed that sporadic cases and infrequent outbreaks were due to either chronic biliary carriers or importations, but this was difficult to characterize with low resolution bacterial typing methods. In Santiago, Chile, typhoid fever persisted at hyperendemic levels through the 1980s until organized control efforts in the 1980s and changes to wastewater policy in 1991 caused annual typhoid incidence to plummet. In this study, we used whole genome sequencing (WGS) to investigate whether recent sporadic cases occurring in Santiago in the 2010s were genomically similar to S. Typhi circulating in the 1980s, or dissimilar, possibly representing importations of S. Typhi from outside of Chile. We found concordance amongst S. Typhi genotypes between the 1980s and 2010s, and differences from genotypes circulating in Southeast Asia and Africa where typhoid remains hyperendemic. Our findings suggest that a proportion of modern, rare typhoid cases in Santiago are autochthonous, and that chronic carriers or another unknown reservoir likely contribute. Broadly, our findings corroborate the epidemiologic importance of long-term reservoirs of typhoid fever decades after typhoid elimination.
Collapse
Affiliation(s)
- Mailis Maes
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Michael J. Sikorski
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Megan E. Carey
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ellen E. Higginson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Zoe A. Dyson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Infectious Diseases, Monash University, Melbourne, Australia
- London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Alda Fernandez
- Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Pamela Araya
- Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Rosanna Lagos
- Centro para Vacunas en Desarollo-Chile (CVD-Chile), Hospital de Niños Roberto del Rio, Santiago, Chile
| | - Juan Carlos Hormazábal
- Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile (ISP), Santiago, Chile
| | - Myron M. Levine
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
47
|
Abstract
Typhoid fever is caused primarily by the enteric microbe Salmonella enterica serovar Typhi and remains a major global health problem with approximately 14 million new infections and 136,000 fatalities annually. While there are antibiotic options available to treat the disease, the global increase in multidrug-resistant strains necessitates alternative therapeutic options. Host-targeted therapeutics present a promising anti-infective strategy against intracellular bacterial pathogens. A cell-based assay identified a compound that inhibits Salmonella proliferation in infected cells, 2-(3-hydroxypropyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione (KH-1), which is devoid of direct activity against Salmonella. The compound inhibits the growth of both antibiotic-sensitive and -resistant Salmonella strains inside macrophages and reduces lactate dehydrogenase (LDH) release from Salmonella-infected cells. Subsequent screening of KH-1 commercial analogs identified 2-(4-fluorobenzyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c] pyrrole-3,9-dione (KH-1-2), which is more effective in controlling Salmonella growth inside macrophages. In vitro KH-1-2 treatment of Salmonella infection resulted in an 8- to 10-fold reduction in bacterial load in infected macrophages. In combination with suboptimal ciprofloxacin treatment, KH-1-2 further reduces Salmonella growth inside macrophages. The toxicity and efficacy of KH-1-2 in controlling Salmonella infection were examined in vivo using a mouse model of typhoid fever. No significant compound-related clinical signs and histological findings of the liver, spleen, or kidney were observed from uninfected mice that were intraperitoneally treated with KH-1-2. KH-1-2 significantly protected mice from a lethal dose of infection by an antibiotic-resistant Salmonella strain. Thus, our study provides support that this is a promising lead compound for the development of a novel host-targeted therapeutic agent to control typhoid fever. IMPORTANCESalmonella spp. cause significant morbidity and mortality worldwide. Typhoidal spp. (e.g., S. Typhi) cause a systemic disease typically treated with antibiotics. However, growing antibiotic resistance is resulting in increased treatment failures. We screened a compound library for those that would reduce Salmonella-induced macrophage toxicity, identifying compound KH-1. KH-1 has no direct effects on the bacteria but limits Salmonella survival in macrophages and protects against lethal infection in a mouse model of typhoid fever. A suboptimal concentration of ciprofloxacin worked in conjunction with the compound to further decrease Salmonella survival in macrophages. An analog (KH-1-2) was identified that possessed increased activity in vitro in macrophages and in vivo against both antibiotic-sensitive and -resistant strains. Thus, we report the identification of a lead compound that may be a useful scaffold as a host-directed antimicrobial against typhoid fever.
Collapse
|
48
|
Feng Y, Wang Z, Chien KY, Chen HL, Liang YH, Hua X, Chiu CH. "Pseudo-pseudogenes" in bacterial genomes: Proteogenomics reveals a wide but low protein expression of pseudogenes in Salmonella enterica. Nucleic Acids Res 2022; 50:5158-5170. [PMID: 35489061 PMCID: PMC9122581 DOI: 10.1093/nar/gkac302] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/03/2022] Open
Abstract
Pseudogenes (genes disrupted by frameshift or in-frame stop codons) are ubiquitously present in the bacterial genome and considered as nonfunctional fossil. Here, we used RNA-seq and mass-spectrometry technologies to measure the transcriptomes and proteomes of Salmonella enterica serovars Paratyphi A and Typhi. All pseudogenes’ mRNA sequences remained disrupted, and were present at comparable levels to their intact homologs. At the protein level, however, 101 out of 161 pseudogenes suggested successful translation, with their low expression regardless of growth conditions, genetic background and pseudogenization causes. The majority of frameshifting detected was compensatory for -1 frameshift mutations. Readthrough of in-frame stop codons primarily involved UAG; and cytosine was the most frequent base adjacent to the codon. Using a fluorescence reporter system, fifteen pseudogenes were confirmed to express successfully in vivo in Escherichia coli. Expression of the intact copy of the fifteen pseudogenes in S. Typhi affected bacterial pathogenesis as revealed in human macrophage and epithelial cell infection models. The above findings suggest the need to revisit the nonstandard translation mechanism as well as the biological role of pseudogenes in the bacterial genome.
Collapse
Affiliation(s)
- Ye Feng
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zeyu Wang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Kun-Yi Chien
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Republic of China
| | - Hsiu-Ling Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Republic of China
| | - Yi-Hua Liang
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Republic of China
| | - Xiaoting Hua
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Republic of China.,Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Republic of China.,Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Republic of China
| |
Collapse
|
49
|
Krishnamoorthy R, Athinarayanan J, Periyasamy VS, Alshuniaber MA, Alshammari G, Hakeem MJ, Ahmed MA, Alshatwi AA. Antibacterial Mechanisms of Zinc Oxide Nanoparticle against Bacterial Food Pathogens Resistant to Beta-Lactam Antibiotics. Molecules 2022; 27:2489. [PMID: 35458685 PMCID: PMC9032754 DOI: 10.3390/molecules27082489] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/24/2022] [Accepted: 04/08/2022] [Indexed: 12/22/2022] Open
Abstract
The increase in β-lactam-resistant Gram-negative bacteria is a severe recurrent problem in the food industry for both producers and consumers. The development of nanotechnology and nanomaterial applications has transformed many features in food science. The antibacterial activity of zinc oxide nanoparticles (ZnO NPs) and their mechanism of action on β-lactam-resistant Gram-negative food pathogens, such as Escherichia coli, Pseudomonas aeruginosa, Salmonella typhi, Serratia marcescens, Klebsiella pneumoniae, and Proteus mirabilis, are investigated in the present paper. The study results demonstrate that ZnO NPs possesses broad-spectrum action against these β-lactamase-producing strains. The minimal inhibitory and minimal bactericidal concentrations vary from 0.04 to 0.08 and 0.12 to 0.24 mg/mL, respectively. The ZnO NPs elevate the level of reactive oxygen species (ROS) and malondialdehyde in the bacterial cells as membrane lipid peroxidation. It has been confirmed from the transmission electron microscopy image of the treated bacterial cells that ZnO NPs diminish the permeable membrane, denature the intracellular proteins, cause DNA damage, and cause membrane leakage. Based on these findings, the action of ZnO NPs has been attributed to the fact that broad-spectrum antibacterial action against β-lactam-resistant Gram-negative food pathogens is mediated by Zn2+ ion-induced oxidative stress, actions via lipid peroxidation and membrane damage, subsequently resulting in depletion, leading to β-lactamase enzyme inhibition, intracellular protein inactivation, DNA damage, and eventually cell death. Based on the findings of the present study, ZnO NPs can be recommended as potent broad-spectrum antibacterial agents against β-lactam-resistant Gram-negative pathogenic strains.
Collapse
Affiliation(s)
- Rajapandiyan Krishnamoorthy
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia or (J.A.); or (V.S.P.); (M.A.A.)
| | - Jegan Athinarayanan
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia or (J.A.); or (V.S.P.); (M.A.A.)
| | - Vaiyapuri Subbarayan Periyasamy
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia or (J.A.); or (V.S.P.); (M.A.A.)
| | - Mohammad A. Alshuniaber
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia or (J.A.); or (V.S.P.); (M.A.A.)
| | - Ghedeir Alshammari
- Department of Food and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia; (G.A.); (M.J.H.); (M.A.A.)
| | - Mohammed Jamal Hakeem
- Department of Food and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia; (G.A.); (M.J.H.); (M.A.A.)
| | - Mohammed Asif Ahmed
- Department of Food and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia; (G.A.); (M.J.H.); (M.A.A.)
| | - Ali A. Alshatwi
- Nanobiotechnology and Molecular Biology Research Lab, Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11541, Saudi Arabia or (J.A.); or (V.S.P.); (M.A.A.)
| |
Collapse
|
50
|
Fang Z, Méresse S. Endomembrane remodeling and dynamics in Salmonella infection. MICROBIAL CELL (GRAZ, AUSTRIA) 2022; 9:24-41. [PMID: 35127930 PMCID: PMC8796136 DOI: 10.15698/mic2022.02.769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022]
Abstract
Salmonellae are bacteria that cause moderate to severe infections in humans, depending on the strain and the immune status of the infected host. These pathogens have the particularity of residing in the cells of the infected host. They are usually found in a vacuolar compartment that the bacteria shape with the help of effector proteins. Following invasion of a eukaryotic cell, the bacterial vacuole undergoes maturation characterized by changes in localization, composition and morphology. In particular, membrane tubules stretching over the microtubule cytoskeleton are formed from the bacterial vacuole. Although these tubules do not occur in all infected cells, they are functionally important and promote intracellular replication. This review focuses on the role and significance of membrane compartment remodeling observed in infected cells and the bacterial and host cell pathways involved.
Collapse
Affiliation(s)
- Ziyan Fang
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|