1
|
Hotinger JA, Campbell IW, Hullahalli K, Osaki A, Waldor MK. Quantification of Salmonella enterica serovar Typhimurium Population Dynamics in Murine Infection Using a Highly Diverse Barcoded Library. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601246. [PMID: 38979326 PMCID: PMC11230369 DOI: 10.1101/2024.06.28.601246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Murine models are often used to study the pathogenicity and dissemination of the enteric pathogen Salmonella enterica serovar Typhimurium. Here, we quantified S. Typhimurium population dynamics in mice using the STAMPR analytic pipeline and a highly diverse S. Typhimurium barcoded library containing ~55,000 unique strains distinguishable by genomic barcodes by enumerating S. Typhimurium founding populations and deciphering routes of spread in mice. We found that a severe bottleneck allowed only one in a million cells from an oral inoculum to establish a niche in the intestine. Furthermore, we observed compartmentalization of pathogen populations throughout the intestine, with few barcodes shared between intestinal segments and feces. This severe bottleneck widened and compartmentalization was reduced after streptomycin treatment, suggesting the microbiota plays a key role in restricting the pathogen's colonization and movement within the intestine. Additionally, there was minimal sharing between the intestine and extraintestinal organ populations, indicating dissemination to extraintestinal sites occurs rapidly, before substantial pathogen expansion in the intestine. Bypassing the intestinal bottleneck by inoculating mice via intravenous or intraperitoneal injection revealed that Salmonella re-enters the intestine after establishing niches in extraintestinal sites by at least two distinct pathways. One pathway results in a diverse intestinal population. The other re-seeding pathway is through the bile, where the pathogen is often clonal, leading to clonal intestinal populations and correlates with gallbladder pathology. Together, these findings deepen our understanding of Salmonella population dynamics.
Collapse
Affiliation(s)
- Julia A. Hotinger
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Ian W. Campbell
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Akina Osaki
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
2
|
Wang X, Sun Y, Guo H, Yan X, Ma S, Yang B, Jiang L. Xylose utilization promotes Salmonella replication within macrophages and systemic infection in mice. Virulence 2024; 15:2435381. [PMID: 39603596 DOI: 10.1080/21505594.2024.2435381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/03/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
The intracellular pathogen Salmonella can cause systemic diseases via its survival and replication in host macrophages. Xylose is the second most abundant sugar in nature and Salmonella can use xylose as its sole carbon source for growth. However, whether xylose utilization contributes to the pathogenicity and intracellular growth of Salmonella has not yet been determined. In this study, we observed that the xylose concentration in macrophages increased during Salmonella infection. Moreover, there was an increase in expression of Salmonella xylose catabolic genes (xylA and xylB) and the transcriptional regulatory gene of xylose metabolism (xylR) in macrophages, revealing the possibility of using host-accumulated xylose by Salmonella for intracellular growth. Mutation of either xylAB or xylR reduced Salmonella replication in macrophages and attenuated the colonization of mouse systemic loci (e.g. the liver and spleen), indicating that xylose utilization promotes Salmonella replication within macrophages and systemic infection in mice. Moreover, we found that xylose utilization by intracellular Salmonella was activated by the cAMP-CRP complex upon detection of low glucose levels in the infected macrophages. Collectively, these findings reveal that although the available glucose decreases during infection, Salmonella can use xylose, which accumulates in infected macrophages, as an alternative carbon source to promote intracellular replication and virulence.
Collapse
Affiliation(s)
- Xinyue Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Yuyang Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Houliang Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Xiaolin Yan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuai Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Bin Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Lingyan Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| |
Collapse
|
3
|
Giorgio RT, Helaine S. Antibiotic-recalcitrant Salmonella during infection. Nat Rev Microbiol 2024:10.1038/s41579-024-01124-z. [PMID: 39558126 DOI: 10.1038/s41579-024-01124-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
Antibiotic-recalcitrant infections, defined as the prolonged carriage of pathogenic bacteria even in the presence of antibiotics, are often caused by bacteria that are genetically susceptible to the drug. These recalcitrant bacteria fail to proliferate in the presence of antibiotics but remain viable such that they may recolonize their niche following antibiotic withdrawal. Significant progress has been made in our understanding of antibiotic-recalcitrant Salmonella, which are thought to be the source of infection relapse. In recent years, it has been shown that recalcitrant bacteria manipulate host immune defences and could directly contribute to the spread of antimicrobial resistance. In this Review, we provide an overview of what is currently known about the antibiotic recalcitrance of Salmonella during infection and highlight knowledge gaps requiring additional research in the future.
Collapse
Affiliation(s)
- Rachel T Giorgio
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
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
|
Girón-Pérez DA, Espinoza-Gonzalez HD, Murillo Cisneros JA, Covantes-Rosales CE, Toledo-Ibarra GA, Díaz-Resendiz KJG, Barcelos-García RG, Benitez-Trinidad AB, Girón-Pérez MI. Diazoxon exposure increases susceptibility to infection by Salmonella Typhimurium. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024:1-12. [PMID: 38842028 DOI: 10.1080/09603123.2024.2363475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
Various exogenous factors, such as microbiological and chemical contamination condition food security. Salmonella Typhimurium (S. Typhimurium) is the cause of salmonellosis. This bacterium utilizes phagocytosis to create bacterial reservoirs. On the other hand, exposure to chemical contaminants, such as pesticides, increases susceptibility to numerous infections. Therefore, this research aims to evaluate the effect of co-exposure to diazoxon and S. Typhimurium on the in vitro infection dynamics. For this purpose, human mononuclear cells were pre-exposed in vitro to diazoxon and then challenged with S. Typhimurium at 1, 8, and 24 h. Bacterial internalization, actin polymerization, and reactive oxygen species (ROS) were analyzed. Obtained data show that mononuclear cells previously exposed to diazoxon exhibit greater internalization of S. Typhimurium. Likewise, greater ROS production and an increase in actin polymerization were observed. Therefore, in the proposed scenario, obtained data suggest that co-exposure to diazoxon and S. Typhimurium increases susceptibility to acquiring an illness.
Collapse
Affiliation(s)
- Daniel Alberto Girón-Pérez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | | | | | - Carlos Eduardo Covantes-Rosales
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Gladys Alejandra Toledo-Ibarra
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Karina Janice Guadalupe Díaz-Resendiz
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Rocío Guadalupe Barcelos-García
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Alma Betsaida Benitez-Trinidad
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| | - Manuel Iván Girón-Pérez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA)-Nayarit, Universidad Autónoma de Nayarit, Tepic, Nayarit, México
- Licenciatura en Biomedicina Ambiental Traslacional (LIBAT), Universidad Autónoma de Nayarit, Tepic, Nayarit, México
| |
Collapse
|
6
|
Dai Y, Zhang M, Liu X, Sun T, Qi W, Ding W, Chen Z, Zhang P, Liu R, Chen H, Chen S, Wang Y, Yue Y, Song N, Wang W, Jia H, Ma Z, Li C, Chen Q, Li B. Salmonella manipulates macrophage migration via SteC-mediated myosin light chain activation to penetrate the gut-vascular barrier. EMBO J 2024; 43:1499-1518. [PMID: 38528181 PMCID: PMC11021425 DOI: 10.1038/s44318-024-00076-7] [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: 07/30/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
The intestinal pathogen Salmonella enterica rapidly enters the bloodstream after the invasion of intestinal epithelial cells, but how Salmonella breaks through the gut-vascular barrier is largely unknown. Here, we report that Salmonella enters the bloodstream through intestinal CX3CR1+ macrophages during early infection. Mechanistically, Salmonella induces the migration/invasion properties of macrophages in a manner dependent on host cell actin and on the pathogen effector SteC. SteC recruits host myosin light chain protein Myl12a and phosphorylates its Ser19 and Thr20 residues. Myl12a phosphorylation results in actin rearrangement, and enhanced migration and invasion of macrophages. SteC is able to utilize a wide range of NTPs other than ATP to phosphorylate Myl12a. We further solved the crystal structure of SteC, which suggests an atypical dimerization-mediated catalytic mechanism. Finally, in vivo data show that SteC-mediated cytoskeleton manipulation is crucial for Salmonella breaching the gut vascular barrier and spreading to target organs.
Collapse
Affiliation(s)
- Yuanji Dai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Min Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoyu Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ting Sun
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wenqi Qi
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Ding
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhe Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Ping Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruirui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Huimin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Siyan Chen
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuzhen Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yingying Yue
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Nannan Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haihong Jia
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongrui Ma
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Cuiling Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| | - Bingqing Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
- Key Lab for Biotech-Drugs of National Health Commission, Jinan, 250117, China.
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, China.
| |
Collapse
|
7
|
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
|
8
|
Wang X, Yang B, Ma S, Yan X, Ma S, Sun H, Sun Y, Jiang L. Lactate promotes Salmonella intracellular replication and systemic infection via driving macrophage M2 polarization. Microbiol Spectr 2023; 11:e0225323. [PMID: 37796020 PMCID: PMC10715217 DOI: 10.1128/spectrum.02253-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE The important enteropathogen Salmonella can cause lethal systemic infection via survival and replication in host macrophages. Lactate represents an abundant intracellular metabolite during bacterial infection, which can also induce macrophage M2 polarization. In this study, we found that macrophage-derived lactate promotes the intracellular replication and systemic infection of Salmonella. During Salmonella infection, lactate via the Salmonella type III secretion system effector SteE promotes macrophage M2 polarization, and the induction of macrophage M2 polarization by lactate is responsible for lactate-mediated Salmonella growth promotion. This study highlights the complex interactions between Salmonella and macrophages and provides an additional perspective on host-pathogen crosstalk at the metabolic interface.
Collapse
Affiliation(s)
- Xinyue Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Bin Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuangshuang Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- Department of Biopharmaceuticals, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaolin Yan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuai Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Hongmin Sun
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Yuyang Sun
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Lingyan Jiang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Thurston TLM, Holden DW. The Salmonella Typhi SPI-2 injectisome enigma. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001405. [PMID: 37862087 PMCID: PMC10634361 DOI: 10.1099/mic.0.001405] [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: 08/29/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
The Salmonella pathogenicity island 2 (SPI-2)-encoded type III secretion system (injectisome) is assembled following uptake of bacteria into vacuoles in mammalian cells. The injectisome translocates virulence proteins (effectors) into infected cells. Numerous studies have established the requirement for a functional SPI-2 injectisome for growth of Salmonella Typhimurium in mouse macrophages, but the results of similar studies involving Salmonella Typhi and human-derived macrophages are not consistent. It is important to clarify the functions of the S. Typhi SPI-2 injectisome, not least because an inactivated SPI-2 injectisome forms the basis for live attenuated S. Typhi vaccines that have undergone extensive trials in humans. Intracellular expression of injectisome genes and effector delivery take longer in the S. Typhi/human macrophage model than for S. Typhimurium and we propose that this could explain the conflicting results. Furthermore, strains of both S. Typhimurium and S. Typhi contain intact genes for several 'core' effectors. In S. Typhimurium these cooperate to regulate the vacuole membrane and contribute to intracellular bacterial replication; similar functions are therefore likely in S. Typhi.
Collapse
Affiliation(s)
- Teresa L. M. Thurston
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, SW7 2AZ, UK
| | - David W. Holden
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
10
|
Turner M, Van Hulzen L, Peta V, Pietri JE. Survival of Salmonella Typhimurium in the hemolymph of the German cockroach vector is limited by both humoral immune factors and hemocytes but not by trehalose metabolism. JOURNAL OF MEDICAL ENTOMOLOGY 2023; 60:875-883. [PMID: 37348971 PMCID: PMC10496438 DOI: 10.1093/jme/tjad076] [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: 02/19/2023] [Revised: 05/15/2023] [Accepted: 06/08/2023] [Indexed: 06/24/2023]
Abstract
The German cockroach (Blattella germanica) has been linked to transmission of Salmonella enterica serovar Typhimurium (S. Typhimurium), but infection dynamics within this vector are poorly characterized. Our recent work has focused on S. Typhimurium infection in the cockroach gut. However, microbial dissemination to the hemolymph is an essential aspect of many vector-borne pathogen transmission cycles and could potentially contribute to S. Typhimurium colonization of cockroaches. Therefore, the goal of this study was to examine the ability of S. Typhimurium to disseminate, survive, and proliferate in the hemolymph of cockroaches after oral infection. We detected only low numbers of bacteria in the hemolymph of a minority of insects (~26%) after oral infection. Further, S. Typhimurium was unable to survive overnight in cell-free hemolymph. Several hypotheses to explain the inability of S. Typhimurium to colonize hemolymph were tested. First, we investigated the ability of S. Typhimurium to metabolize trehalose, the primary sugar in hemolymph. S. Typhimurium grew efficiently in vitro using trehalose as a sole carbon source and mutant strains lacking trehalose metabolism genes exhibited no growth deficiencies in media mimicking the composition of hemolymph, suggesting that trehalose metabolism ability is not a factor involved in restricting survival in hemolymph. On the other hand, heat-inactivated cell-free hemolymph was permissive of S. Typhimurium growth, demonstrating that survival in hemolymph is limited specifically by heat-labile humoral factors. The involvement of cellular immune responses was also investigated and cockroach hemocytes in culture were observed to internalize S. Typhimurium within 1 h of exposure. Most hemocytes harbored few to no bacteria after 24 h, indicating that hemocyte responses are additionally involved in clearing infection from the hemolymph. However, dense intracellular clusters of S. Typhimurium were observed sporadically, suggesting a small subset of hemocytes may serve as reservoirs for bacterial replication. Together, our results reveal that a minute proportion of ingested S. Typhimurium is able to escape the cockroach gut and enter the hemolymph, but this systemic population is limited by both humoral effectors and hemocytes. Thus, we conclude that invasion of the hemolymph appears minimally important for colonization of the cockroach vector and that colonization of the gut is the main driver of vector-borne transmission. Our insight into the antimicrobial mechanisms of cockroach hemolymph also highlights the strong ability of these prevalent pests/vectors to cope with frequent infectious challenges in septic habitats.
Collapse
Affiliation(s)
- Matthew Turner
- Sanford School of Medicine, Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA
| | - Landen Van Hulzen
- Sanford School of Medicine, Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA
| | - Vincent Peta
- Sanford School of Medicine, Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA
| | - Jose E Pietri
- Sanford School of Medicine, Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
11
|
Chatterjee R, Chaudhuri D, Setty SRG, Chakravortty D. Deceiving the big eaters: Salmonella Typhimurium SopB subverts host cell xenophagy in macrophages via dual mechanisms. Microbes Infect 2023; 25:105128. [PMID: 37019426 DOI: 10.1016/j.micinf.2023.105128] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Salmonella, a stealthy facultative intracellular pathogen, utilises an array of host immune evasion strategies. This facilitates successful survival via replicative niche establishment in otherwise hostile environments such as macrophages. Salmonella survives in and utilises macrophages for effective dissemination, ultimately leading to systemic infection. Bacterial xenophagy or macro-autophagy is an important host defense mechanism in macrophages. Here, we report for the first time that the Salmonella pathogenicity island-1 (SPI-1) effector SopB is involved in subverting host autophagy via dual mechanisms. SopB is a phosphoinositide phosphatase capable of altering the phosphoinositide dynamics of the host cell. Here, we demonstrate that SopB mediates escape from autophagy by inhibiting the terminal fusion of Salmonella-containing vacuoles (SCVs) with lysosomes and/or autophagosomes. We also report that SopB downregulates overall lysosomal biogenesis by modulating the Akt-transcription factor EB (TFEB) axis via restricting the latter's nuclear localisation. TFEB is a master regulator of lysosomal biogenesis and autophagy. This reduces the overall lysosome content inside host macrophages, further facilitating the survival of Salmonella in macrophages and systemic dissemination of Salmonella.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Debalina Chaudhuri
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Subba Rao Gangi Setty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru, India; Indian Institute of Science Research and Education, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
12
|
Engel S, Bachem A, Strugnell RA, Strasser A, Herold MJ, Bedoui S. Functional flexibility and plasticity in immune control of systemic Salmonella infection. Curr Opin Immunol 2023; 83:102343. [PMID: 37245415 DOI: 10.1016/j.coi.2023.102343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/30/2023]
Abstract
Immunity to systemic Salmonella infection depends on multiple effector mechanisms. Lymphocyte-derived interferon gamma (IFN-γ) enhances cell-intrinsic bactericidal capabilities to antagonize the hijacking of phagocytes as replicative niches for Salmonella. Programmed cell death (PCD) provides another means through which phagocytes fight against intracellular Salmonella. We describe remarkable levels of flexibility with which the host coordinates and adapts these responses. This involves interchangeable cellular sources of IFN-γ regulated by innate and adaptive cues, and the rewiring of PCD pathways in previously unknown ways. We discuss that such plasticity is likely the consequence of host-pathogen coevolution and raise the possibility of further functional overlap between these seemingly distinct processes.
Collapse
Affiliation(s)
- Sven Engel
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Richard A Strugnell
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Jiang L, Li W, Hou X, Ma S, Wang X, Yan X, Yang B, Huang D, Liu B, Feng L. Nitric oxide is a host cue for Salmonella Typhimurium systemic infection in mice. Commun Biol 2023; 6:501. [PMID: 37161082 PMCID: PMC10169850 DOI: 10.1038/s42003-023-04876-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 04/26/2023] [Indexed: 05/11/2023] Open
Abstract
Nitric oxide (NO) is produced as an innate immune response against microbial infections. Salmonella Typhimurium (S. Typhimurium), the major causative pathogen of human gastroenteritis, induces more severe systemic disease in mice. However, host factors contributing to the difference in species-related virulence are unknown. Here, we report that host NO production promotes S. Typhimurium replication in mouse macrophages at the early infection stage by activating Salmonella pathogenicity island-2 (SPI-2). The NO signaling-induced SPI-2 activation is mediated by Fnr and PhoP/Q two-component system. NO significantly induced fnr transcription, while Fnr directly activated phoP/Q transcription. Mouse infection assays revealed a NO-dependent increase in bacterial burden in systemic organs during the initial days of infection, indicating an early contribution of host NO to virulence. This study reveals a host signaling-mediated virulence activation pathway in S. Typhimurium that contributes significantly to its systemic infection in mice, providing further insights into Salmonella pathogenesis and host-pathogen interaction.
Collapse
Affiliation(s)
- Lingyan Jiang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Wanwu Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xi Hou
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Shuai Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xinyue Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xiaolin Yan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Bin Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Di Huang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Bin Liu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Lu Feng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China.
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China.
| |
Collapse
|
14
|
Abstract
The major function of the mammalian immune system is to prevent and control infections caused by enteropathogens that collectively have altered human destiny. In fact, as the gastrointestinal tissues are the major interface of mammals with the environment, up to 70% of the human immune system is dedicated to patrolling them The defenses are multi-tiered and include the endogenous microflora that mediate colonization resistance as well as physical barriers intended to compartmentalize infections. The gastrointestinal tract and associated lymphoid tissue are also protected by sophisticated interleaved arrays of active innate and adaptive immune defenses. Remarkably, some bacterial enteropathogens have acquired an arsenal of virulence factors with which they neutralize all these formidable barriers to infection, causing disease ranging from mild self-limiting gastroenteritis to in some cases devastating human disease.
Collapse
Affiliation(s)
- Micah J. Worley
- Department of Biology, University of Louisville, Louisville, Kentucky, USA,CONTACT Micah J. Worley Department of Biology, University of Louisville, 139 Life Sciences Bldg, Louisville, Kentucky, USA
| |
Collapse
|
15
|
Shikov AE, Belousova ME, Belousov MV, Nizhnikov AA, Antonets KS. Salmonella-Based Biorodenticides: Past Applications and Current Contradictions. Int J Mol Sci 2022; 23:ijms232314595. [PMID: 36498920 PMCID: PMC9736839 DOI: 10.3390/ijms232314595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/03/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
The idea of using pathogens to control pests has existed since the end of the 19th century. Enterobacteria from the genus Salmonella, discovered at that time, are the causative agents of many serious diseases in mammals often leading to death. Mostly, the strains of Salmonella are able to infect a wide spectrum of hosts belonging to vertebrates, but some of them show host restriction. Several strains of these bacteria have been used as biorodenticides due to the host restriction until they were banned in many countries in the second part of the 20th century. The main reason for the ban was their potential pathogenicity for some domestic animals and poultry and the outbreaks of gastroenteritis in humans. Since that time, a lot of data regarding the host specificity and host restriction of different strains of Salmonella have been accumulated, and the complexity of the molecular mechanisms affecting it has been uncovered. In this review, we summarize the data regarding the history of studying and application of Salmonella-based rodenticides, discuss molecular systems controlling the specificity of Salmonella interactions within its multicellular hosts at different stages of infection, and attempt to reconstruct the network of genes and their allelic variants which might affect the host-restriction mechanisms.
Collapse
Affiliation(s)
- Anton E. Shikov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Maria E. Belousova
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
| | - Mikhail V. Belousov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Anton A. Nizhnikov
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Kirill S. Antonets
- Laboratory for Proteomics of Supra-Organismal Systems, All-Russia Research Institute for Agricultural Microbiology (ARRIAM), Pushkin, St. Petersburg 196608, Russia
- Faculty of Biology, St. Petersburg State University, St. Petersburg 199034, Russia
- Correspondence:
| |
Collapse
|
16
|
Lin HC, Wu YL, Hsu CY, Lin MY, Chen LH, Shiau CW, Chiu HC. Discovery of antipsychotic loxapine derivatives against intracellular multidrug-resistant bacteria. RSC Med Chem 2022; 13:1361-1366. [PMID: 36439974 PMCID: PMC9667769 DOI: 10.1039/d2md00182a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/02/2022] [Indexed: 08/13/2023] Open
Abstract
The emergence and spread of multidrug-resistant bacteria highlight the need for new antibacterial interventions. A screening of 24 newly synthesized dibenzoxazepines identified a small molecule compound, SW14, with potent inhibitory activity against intracellular multidrug-resistant and fluoroquinolone-resistant strains of S. typhimurium in macrophages and epithelial cells. Moreover, intra-macrophagic Salmonella typhi, Yersinia enterocolitica, and Listeria monocytogenes and methicillin-resistant Staphylococcus aureus are also susceptible to SW14. Overall, our findings suggest that SW14 has a broad-spectrum activity against intracellular bacteria.
Collapse
Affiliation(s)
- Hsueh-Chun Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan
| | - Yi-Lun Wu
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 11221 Taiwan
| | - Cheng-Yun Hsu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan
| | - Man-Yi Lin
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 11221 Taiwan
| | - Ling-Han Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 11221 Taiwan
| | - Hao-Chieh Chiu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei 10021 Taiwan
| |
Collapse
|
17
|
Ménard S, Lacroix-Lamandé S, Ehrhardt K, Yan J, Grassl GA, Wiedemann A. Cross-Talk Between the Intestinal Epithelium and Salmonella Typhimurium. Front Microbiol 2022; 13:906238. [PMID: 35733975 PMCID: PMC9207452 DOI: 10.3389/fmicb.2022.906238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Salmonella enterica serovars are invasive gram-negative bacteria, causing a wide range of diseases from gastroenteritis to typhoid fever, representing a public health threat around the world. Salmonella gains access to the intestinal lumen after oral ingestion of contaminated food or water. The crucial initial step to establish infection is the interaction with the intestinal epithelium. Human-adapted serovars such as S. Typhi or S. Paratyphi disseminate to systemic organs and induce life-threatening disease known as typhoid fever, whereas broad-host serovars such as S. Typhimurium usually are limited to the intestine and responsible for gastroenteritis in humans. To overcome intestinal epithelial barrier, Salmonella developed mechanisms to induce cellular invasion, intracellular replication and to face host defence mechanisms. Depending on the serovar and the respective host organism, disease symptoms differ and are linked to the ability of the bacteria to manipulate the epithelial barrier for its own profit and cross the intestinal epithelium.This review will focus on S. Typhimurium (STm). To better understand STm pathogenesis, it is crucial to characterize the crosstalk between STm and the intestinal epithelium and decipher the mechanisms and epithelial cell types involved. Thus, the purpose of this review is to summarize our current knowledge on the molecular dialogue between STm and the various cell types constituting the intestinal epithelium with a focus on the mechanisms developed by STm to cross the intestinal epithelium and access to subepithelial or systemic sites and survive host defense mechanisms.
Collapse
Affiliation(s)
- Sandrine Ménard
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | | | - Katrin Ehrhardt
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Jin Yan
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
- Research Center of Digestive Disease, Central South University, Changsha, China
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Agnès Wiedemann
- IRSD - Institut de Recherche en Santé Digestive, Université́ de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- *Correspondence: Agnès Wiedemann,
| |
Collapse
|
18
|
Devlin JR, Santus W, Mendez J, Peng W, Yu A, Wang J, Alejandro-Navarreto X, Kiernan K, Singh M, Jiang P, Mechref Y, Behnsen J. Salmonella enterica serovar Typhimurium chitinases modulate the intestinal glycome and promote small intestinal invasion. PLoS Pathog 2022; 18:e1010167. [PMID: 35482787 PMCID: PMC9049507 DOI: 10.1371/journal.ppat.1010167] [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: 12/03/2021] [Accepted: 03/23/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is one of the leading causes of food-borne illnesses worldwide. To colonize the gastrointestinal tract, S. Typhimurium produces multiple virulence factors that facilitate cellular invasion. Chitinases have been recently emerging as virulence factors for various pathogenic bacterial species, and the S. Typhimurium genome contains two annotated chitinases: STM0018 (chiA) and STM0233. However, the role of these chitinases during S. Typhimurium pathogenesis is unknown. The putative chitinase STM0233 has not been studied previously, and only limited data exists on ChiA. Chitinases typically hydrolyze chitin polymers, which are absent in vertebrates. However, chiA expression was detected in infection models and purified ChiA cleaved carbohydrate subunits present on mammalian surface glycoproteins, indicating a role during pathogenesis. Here, we demonstrate that expression of chiA and STM0233 is upregulated in the mouse gut and that both chitinases facilitate epithelial cell adhesion and invasion. S. Typhimurium lacking both chitinases showed a 70% reduction in invasion of small intestinal epithelial cells in vitro. In a gastroenteritis mouse model, chitinase-deficient S. Typhimurium strains were also significantly attenuated in the invasion of small intestinal tissue. This reduced invasion resulted in significantly delayed S. Typhimurium dissemination to the spleen and the liver, but chitinases were not required for systemic survival. The invasion defect of the chitinase-deficient strain was rescued by the presence of wild-type S. Typhimurium, suggesting that chitinases are secreted. By analyzing N-linked glycans of small intestinal cells, we identified specific N-acetylglucosamine-containing glycans as potential extracellular targets of S. Typhimurium chitinases. This analysis also revealed a differential abundance of Lewis X/A-containing glycans that is likely a result of host cell modulation due to the detection of S. Typhimurium chitinases. Similar glycomic changes elicited by chitinase deficient strains indicate functional redundancy of the chitinases. Overall, our results demonstrate that S. Typhimurium chitinases contribute to intestinal adhesion and invasion through modulation of the host glycome.
Collapse
Affiliation(s)
- Jason R. Devlin
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - William Santus
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Jorge Mendez
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Wenjing Peng
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Aiying Yu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Junyao Wang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Xiomarie Alejandro-Navarreto
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Kaitlyn Kiernan
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Manmeet Singh
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Peilin Jiang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, United States of America
| | - Judith Behnsen
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
19
|
Wellawa DH, Lam PKS, White AP, Gomis S, Allan B, Köster W. High Affinity Iron Acquisition Systems Facilitate but Are Not Essential for Colonization of Chickens by Salmonella Enteritidis. Front Microbiol 2022; 13:824052. [PMID: 35308377 PMCID: PMC8928163 DOI: 10.3389/fmicb.2022.824052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/24/2022] [Indexed: 01/02/2023] Open
Abstract
The roles of TonB mediated Fe3+ (ferric iron) uptake via enterobactin (involving biosynthesis genes entABCDEF) and Fe2+ (ferrous iron) uptake through the FeoABC transporter are poorly defined in the context of chicken-Salmonella interactions. Both uptake systems are believed to be the major contributors of iron supply in the Salmonella life cycle. Current evidence suggests that these iron uptake systems play a major role in pathogenesis in mammals and as such, they represent promising antibacterial targets with therapeutic potential. We investigated the role of these iron uptake mechanisms regarding the ability of Salmonella Enteritidis (SEn) strains to colonize in a chicken infection model. Further we constructed a bioluminescent reporter to sense iron limitation during gastrointestinal colonization of Salmonella in chicken via ex vivo imaging. Our data indicated that there is some redundancy between the ferric and ferrous iron uptake mechanisms regarding iron acquisition during SEn pathogenesis in chicken. We believe that this redundancy of iron acquisition in the host reservoir may be the consequence of adaptation to unique avian environments, and thus warrants further investigation. To our knowledge, this the first report providing direct evidence that both enterobactin synthesis and FeoABC mediated iron uptake contribute to the virulence of SEn in chickens.
Collapse
Affiliation(s)
- Dinesh H Wellawa
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Po-King S Lam
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Aaron P White
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Brenda Allan
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Wolfgang Köster
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
20
|
Troha K, Ayres JS. Cooperative defenses during enteropathogenic infection. Curr Opin Microbiol 2022; 65:123-130. [PMID: 34847524 PMCID: PMC8818259 DOI: 10.1016/j.mib.2021.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023]
Abstract
During their co-evolution with pathogens, hosts acquired defensive health strategies that allow them to maintain their health or promote recovery when challenged with infections. The cooperative defense system is a largely unexplored branch of these evolved defense strategies. Cooperative defenses limit physiological damage and promote health without having a negative impact on a pathogen's ability to survive and replicate within the host. Here, we review recent discoveries in the new field of cooperative defenses using the model pathogens Citrobacter rodentium and Salmonella enterica. We discuss not only host-encoded but also pathogen-encoded mechanisms of cooperative defenses. Cooperative defenses remain an untapped resource in clinical medicine. With a global pandemic exacerbated by a lack of vaccine access and a worldwide rise in antibiotic resistance, the study of cooperative defenses offers an opportunity to safeguard health in the face of pathogenic infection.
Collapse
Affiliation(s)
- Katia Troha
- Molecular and Systems Physiology Lab, Gene Expression Lab, Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, Gene Expression Lab, Nomis Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Correspondence:
| |
Collapse
|
21
|
Stolzer I, Schickedanz L, Chiriac MT, López-Posadas R, Grassl GA, Mattner J, Wirtz S, Winner B, Neurath MF, Günther C. STAT1 coordinates intestinal epithelial cell death during gastrointestinal infection upstream of Caspase-8. Mucosal Immunol 2022; 15:130-142. [PMID: 34497340 PMCID: PMC8732278 DOI: 10.1038/s41385-021-00450-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 02/04/2023]
Abstract
Intestinal homeostasis and the maintenance of the intestinal epithelial barrier are essential components of host defense during gastrointestinal Salmonella Typhimurium infection. Both require a strict regulation of cell death. However, the molecular pathways regulating epithelial cell death have not been completely understood. Here, we elucidated the contribution of central mechanisms of regulated cell death and upstream regulatory components during gastrointestinal infection. Mice lacking Caspase-8 in the intestinal epithelium are highly sensitive towards bacterial induced enteritis and intestinal inflammation, resulting in an enhanced lethality of these mice. This phenotype was associated with an increased STAT1 activation during Salmonella infection. Cell death, barrier breakdown and systemic infection were abrogated by an additional deletion of STAT1 in Casp8ΔIEC mice. In the absence of epithelial STAT1, loss of epithelial cells was abolished which was accompanied by a reduced Caspase-8 activation. Mechanistically, we demonstrate that epithelial STAT1 acts upstream of Caspase-8-dependent as well as -independent cell death and thus might play a major role at the crossroad of several central cell death pathways in the intestinal epithelium. In summary, we uncovered that transcriptional control of STAT1 is an essential host response mechanism that is required for the maintenance of intestinal barrier function and host survival.
Collapse
Affiliation(s)
- Iris Stolzer
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Laura Schickedanz
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Mircea T. Chiriac
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Rocío López-Posadas
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Jochen Mattner
- grid.5330.50000 0001 2107 3311Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Stefan Wirtz
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Beate Winner
- grid.411668.c0000 0000 9935 6525Department of Stem Cell Biology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Center for Rare Diseases Erlangen (ZSEER), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus F. Neurath
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Claudia Günther
- grid.411668.c0000 0000 9935 6525Department of Medicine 1, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany ,grid.411668.c0000 0000 9935 6525Deutsches Zentrum Immuntherapie DZI, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| |
Collapse
|
22
|
Roche SM, Holbert S, Le Vern Y, Rossignol C, Rossignol A, Velge P, Virlogeux-Payant I. A large panel of chicken cells are invaded in vivo by Salmonella Typhimurium even when depleted of all known invasion factors. Open Biol 2021; 11:210117. [PMID: 34784793 PMCID: PMC8596019 DOI: 10.1098/rsob.210117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Poultry are the main source of human infection by Salmonella. As infected poultry are asymptomatic, identifying infected poultry farms is difficult, thus controlling animal infections is of primary importance. As cell tropism is known to govern disease, our aim was therefore to identify infected host-cell types in the organs of chicks known to be involved in Salmonella infection and investigate the role of the three known invasion factors in this process (T3SS-1, Rck and PagN). Chicks were inoculated with wild-type or isogenic fluorescent Salmonella Typhimurium mutants via the intracoelomic route. Our results show that liver, spleen, gall bladder and aortic vessels could be foci of infection, and that phagocytic and non-phagocytic cells, including immune, epithelial and endothelial cells, are invaded in vivo in each organ. Moreover, a mutant defective for the T3SS-1, Rck and PagN remained able to colonize organs like the wild-type strain and invaded non-phagocytic cells in each organ studied. As the infection of the gall bladder had not previously been described in chicks, invasion of gall bladder cells was confirmed by immunohistochemistry and infection was shown to last several weeks after inoculation. Altogether, for the first time these findings provide insights into cell tropism of Salmonella in relevant organs involved in Salmonella infection in chicks and also demonstrate that the known invasion factors are not required for entry into these cell types.
Collapse
Affiliation(s)
- S. M. Roche
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | - S. Holbert
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | - Y. Le Vern
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | - C. Rossignol
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | - A. Rossignol
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | - P. Velge
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France
| | | |
Collapse
|
23
|
Fernandes SE, Saini DK. The ERK-p38MAPK-STAT3 Signalling Axis Regulates iNOS Expression and Salmonella Infection in Senescent Cells. Front Cell Infect Microbiol 2021; 11:744013. [PMID: 34746026 PMCID: PMC8569389 DOI: 10.3389/fcimb.2021.744013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/05/2021] [Indexed: 01/10/2023] Open
Abstract
The cellular changes occurring due to senescence like proliferation arrest, increase in free radical levels, and secretion of pro-inflammatory cytokines have been well studied, but its associated alteration in intracellular signalling networks has been scarcely explored. In this study, we examine the roles of three major kinases viz. p38 MAPK, ERK, and STAT3 in regulating iNOS expression and thereby the levels of the free radical Nitric oxide in senescent cells. Our study revealed that these kinases could differentially regulate iNOS in senescent cells compared to non-senescent cells. Further, we tested the physiological relevance of these alterations with Salmonella infection assays and established an inter-regulatory network between these kinases unique to infected senescent cells. Overall, our findings show how key signalling networks may be rewired in senescent cells rendering them phenotypically different.
Collapse
Affiliation(s)
- Sheryl Erica Fernandes
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Deepak Kumar Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
- Center For BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
24
|
Mancilla-Becerra LM, Orozco-García AG, Armas-Puente P, Martínez-Chávez L, Barba-León J. Pathogenesis of Salmonella Newport to BALB/c mice when internalized into cherry tomatoes. ENVIRONMENTAL MICROBIOLOGY REPORTS 2021; 13:495-502. [PMID: 33543601 DOI: 10.1111/1758-2229.12930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
Salmonella Newport is a serovar frequently associated with outbreaks caused by consumption of raw tomatoes. This study tested the internalization of S. Newport-45 into cherry tomatoes and its resulting pathogenicity in vivo. Pathogenicity of S. Newport-45 was tested in BALB/c mice inoculated orally with either LB grown or cherry tomatoes homogenates internally contaminated with S. Newport-45. CFU of S. Newport-45 was recovered from the gastrointestinal tract, liver and spleen of the inoculated animals. Similar loads (p > 0.05) were recovered from the GI tract of BALB/c mice inoculated with S. Newport-45 grown in LB or with cherry tomato homogenates internally contaminated. Spread of S. Newport-45 to the liver of mice increased (p < 0.05) when they were inoculated with homogenates of cherry tomatoes internally contaminated with S. Newport-45 stored for 3 days compared with bacteria grown in LB. Salmonella Newport-45 hilA and rpoS genes were transcribed when the bacteria were inside the cherry tomato. The results obtained in this study show S. Newport-45 pathogenicity when it is internalized in a raw consumption fruit such as cherry tomato.
Collapse
Affiliation(s)
- Lilia Mercedes Mancilla-Becerra
- Departamento de Salud Pública, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Zapopan, Jalisco, 45200, Mexico
| | - Adriana Guadalupe Orozco-García
- Departamento de Salud Pública, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Zapopan, Jalisco, 45200, Mexico
| | - Priscila Armas-Puente
- Departamento de Salud Pública, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Zapopan, Jalisco, 45200, Mexico
| | - Liliana Martínez-Chávez
- Departamento de Farmacobiología, Universidad de Guadalajara, Boulevard Marcelino García Barragán 1421, esq. Calzada Olímpica, Guadalajara, Jalisco, 44430, Mexico
| | - Jeannette Barba-León
- Departamento de Salud Pública, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Zapopan, Jalisco, 45200, Mexico
| |
Collapse
|
25
|
Analysis of In Vivo Transcriptome of Intracellular Bacterial Pathogen Salmonella enterica serovar Typhmurium Isolated from Mouse Spleen. Pathogens 2021; 10:pathogens10070823. [PMID: 34209260 PMCID: PMC8308634 DOI: 10.3390/pathogens10070823] [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: 06/09/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is an important intracellular pathogen that poses a health threat to humans. This study tries to clarify the mechanism of Salmonella survival and reproduction in the host. In this study, high-throughput sequencing analysis was performed on RNA extracted from the strains isolated from infected mouse spleens and an S. Typhimurium reference strain (ATCC 14028) based on the BGISEQ-500 platform. A total of 1340 significant differentially expressed genes (DEGs) were screened. Functional annotation revealed DEGs associated with regulation, metabolism, transport and binding, pathogenesis, and motility. Through data mining and literature retrieval, 26 of the 58 upregulated DEGs (FPKM > 10) were not reported to be related to the adaptation to intracellular survival and were classified as candidate key genes (CKGs) for survival and proliferation in vivo. Our data contribute to our understanding of the mechanisms used by Salmonella to regulate virulence gene expression whilst replicating inside mammalian cells.
Collapse
|
26
|
Zahid MSH, Varma DM, Johnson MM, Landavazo A, Bachelder EM, Blough BE, Ainslie KM. Overcoming reduced antibiotic susceptibility in intracellular Salmonella enterica serovar Typhimurium using AR-12. FEMS Microbiol Lett 2021; 368:6293843. [PMID: 34089315 DOI: 10.1093/femsle/fnab062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/02/2021] [Indexed: 11/14/2022] Open
Abstract
Host-directed therapies (HDTs) could enhance the activity of traditional antibiotics. AR-12 is a promising HDT against intracellular pathogens including Salmonella enterica serovar Typhimurium, and has been shown to act through modulation of autophagy and the Akt kinase pathway. Since AR-12 does not inhibit the growth of planktonic bacteria but only works in conjunction with the infected host-cell, we hypothesized that AR-12 could enhance the activity of antibiotics in less-susceptible strains in the intracellular host environment. We found that repetitive passaging of S. typhimurium in macrophages in the absence of antibiotics led to a 4-fold reduction in their intracellular susceptibility to streptomycin (STR), but had no effect on the bacteria's sensitivity to AR-12. Moreover, when the host-passaged strains were treated with a combined therapy of AR-12 and STR, there was a significant reduction of intracellular bacterial burden compared to STR monotherapy. Additionally, co-treatment of macrophages infected with multi-drug resistant S. typhimurium with AR-12 and STR or ampicillin showed enhanced clearance of the intracellular bacteria. The drug combination did not elicit this effect on planktonic bacteria. Overall, AR-12 enhanced the clearance of less susceptible S. typhimurium in an intracellular environment.
Collapse
Affiliation(s)
- M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Devika M Varma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Monica M Johnson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Antonio Landavazo
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27709, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,School of Medicine, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| |
Collapse
|
27
|
Fernandes SE, Alakesh A, Rajmani RS, Jhunjhunwala S, Saini DK. Aging associated altered response to intracellular bacterial infections and its implication on the host. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119063. [PMID: 34022281 DOI: 10.1016/j.bbamcr.2021.119063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/10/2023]
Abstract
The effects of senescence on geriatric disorders are well explored, but how it influences infections in the elderly is poorly addressed. Here, we show that several anti-microbial responses are elevated in senescent epithelial cells and old mice, which results in decreased bacterial survival in the host after infection. We identify higher levels of iNOS as a crucial host response and show that p38 MAPK in senescent epithelial cells acts as a negative regulator of iNOS transcription. However, in older mice, the ability to impede bacterial infection does not result in enhanced survival, possibly because elevated pro-inflammatory responses are not countered by a robust host protective anti-inflammatory response. Overall, while addressing an alternate advantage of senescent cells, our study demonstrates that infection-associated morbidity in the elderly may not be the sole outcome of pathogen loads but may also be influenced by the host's ability to resolve inflammation-induced damage.
Collapse
Affiliation(s)
- Sheryl Erica Fernandes
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Alakesh Alakesh
- Center For BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - R S Rajmani
- Center for Infectious Disease Research, Indian Institute of Science, Bangalore 560012, India
| | - Siddharth Jhunjhunwala
- Center For BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Deepak Kumar Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India; Center For BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; Center for Infectious Disease Research, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
28
|
Stolzer I, Ruder B, Neurath MF, Günther C. Interferons at the crossroad of cell death pathways during gastrointestinal inflammation and infection. Int J Med Microbiol 2021; 311:151491. [PMID: 33662871 DOI: 10.1016/j.ijmm.2021.151491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Interferons (IFNs) are pleiotropic immune-modulatory cytokines that are well known for their essential role in host defense against viruses, bacteria, and other pathogenic microorganisms. They can exert both, protective or destructive functions depending on the microorganism, the targeted tissue and the cellular context. Interferon signaling results in the induction of IFN-stimulated genes (ISGs) influencing different cellular pathways including direct anti-viral/anti-bacterial response, immune-modulation or cell death. Multiple pathways leading to host cell death have been described, and it is becoming clear that depending on the cellular context, IFN-induced cell death can be beneficial for both: host and pathogen. Accordingly, activation or repression of corresponding signaling mechanisms occurs during various types of infection but is also an important pathway for gastrointestinal inflammation and tissue damage. In this review, we summarize the role of interferons at the crossroad of various cell death pathways in the gut during inflammation and infection.
Collapse
Affiliation(s)
- Iris Stolzer
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Barbara Ruder
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander-Universität (FAU), Erlangen, Nürnberg, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU), Erlangen, Germany.
| |
Collapse
|
29
|
Machata S, Sreekantapuram S, Hünniger K, Kurzai O, Dunker C, Schubert K, Krüger W, Schulze-Richter B, Speth C, Rambach G, Jacobsen ID. Significant Differences in Host-Pathogen Interactions Between Murine and Human Whole Blood. Front Immunol 2021; 11:565869. [PMID: 33519798 PMCID: PMC7843371 DOI: 10.3389/fimmu.2020.565869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023] Open
Abstract
Murine infection models are widely used to study systemic candidiasis caused by C. albicans. Whole-blood models can help to elucidate host-pathogens interactions and have been used for several Candida species in human blood. We adapted the human whole-blood model to murine blood. Unlike human blood, murine blood was unable to reduce fungal burden and more substantial filamentation of C. albicans was observed. This coincided with less fungal association with leukocytes, especially neutrophils. The lower neutrophil number in murine blood only partially explains insufficient infection and filamentation control, as spiking with murine neutrophils had only limited effects on fungal killing. Furthermore, increased fungal survival is not mediated by enhanced filamentation, as a filament-deficient mutant was likewise not eliminated. We also observed host-dependent differences for interaction of platelets with C. albicans, showing enhanced platelet aggregation, adhesion and activation in murine blood. For human blood, opsonization was shown to decrease platelet interaction suggesting that complement factors interfere with fungus-to-platelet binding. Our results reveal substantial differences between murine and human whole-blood models infected with C. albicans and thereby demonstrate limitations in the translatability of this ex vivo model between hosts.
Collapse
Affiliation(s)
- Silke Machata
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Sravya Sreekantapuram
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Kerstin Hünniger
- Research Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Oliver Kurzai
- Research Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Christine Dunker
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Katja Schubert
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Wibke Krüger
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Bianca Schulze-Richter
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Cornelia Speth
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Rambach
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ilse D. Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
30
|
Eisenreich W, Rudel T, Heesemann J, Goebel W. Persistence of Intracellular Bacterial Pathogens-With a Focus on the Metabolic Perspective. Front Cell Infect Microbiol 2021; 10:615450. [PMID: 33520740 PMCID: PMC7841308 DOI: 10.3389/fcimb.2020.615450] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
Persistence has evolved as a potent survival strategy to overcome adverse environmental conditions. This capability is common to almost all bacteria, including all human bacterial pathogens and likely connected to chronic infections caused by some of these pathogens. Although the majority of a bacterial cell population will be killed by the particular stressors, like antibiotics, oxygen and nitrogen radicals, nutrient starvation and others, a varying subpopulation (termed persisters) will withstand the stress situation and will be able to revive once the stress is removed. Several factors and pathways have been identified in the past that apparently favor the formation of persistence, such as various toxin/antitoxin modules or stringent response together with the alarmone (p)ppGpp. However, persistence can occur stochastically in few cells even of stress-free bacterial populations. Growth of these cells could then be induced by the stress conditions. In this review, we focus on the persister formation of human intracellular bacterial pathogens, some of which belong to the most successful persister producers but lack some or even all of the assumed persistence-triggering factors and pathways. We propose a mechanism for the persister formation of these bacterial pathogens which is based on their specific intracellular bipartite metabolism. We postulate that this mode of metabolism ultimately leads, under certain starvation conditions, to the stalling of DNA replication initiation which may be causative for the persister state.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Department of Chemistry, Chair of Biochemistry, Technische Universität München, Garching, Germany
| | - Thomas Rudel
- Chair of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Jürgen Heesemann
- Max von Pettenkofer-Institute, Ludwig Maximilian University of Munich, München, Germany
| | - Werner Goebel
- Max von Pettenkofer-Institute, Ludwig Maximilian University of Munich, München, Germany
| |
Collapse
|
31
|
Schmidt S, Sassu EL, Vatzia E, Pierron A, Lagler J, Mair KH, Stadler M, Knecht C, Spergser J, Dolezal M, Springer S, Theuß T, Fachinger V, Ladinig A, Saalmüller A, Gerner W. Vaccination and Infection of Swine With Salmonella Typhimurium Induces a Systemic and Local Multifunctional CD4 + T-Cell Response. Front Immunol 2021; 11:603089. [PMID: 33584671 PMCID: PMC7874209 DOI: 10.3389/fimmu.2020.603089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/25/2020] [Indexed: 01/14/2023] Open
Abstract
The gram-negative facultative intracellular bacteria Salmonella Typhimurium (STM) often leads to subclinical infections in pigs, but can also cause severe enterocolitis in this species. Due to its high zoonotic potential, the pathogen is likewise dangerous for humans. Vaccination with a live attenuated STM strain (Salmoporc) is regarded as an effective method to control STM infections in affected pig herds. However, information on the cellular immune response of swine against STM is still scarce. In this study, we investigated the T-cell immune response in pigs that were vaccinated twice with Salmoporc followed by a challenge infection with a virulent STM strain. Blood- and organ-derived lymphocytes (spleen, tonsils, jejunal and ileocolic lymph nodes, jejunum, ileum) were stimulated in vitro with heat-inactivated STM. Subsequently, CD4+ T cells present in these cell preparations were analyzed for the production of IFN-γ, TNF-α, and IL-17A by flow cytometry and Boolean gating. Highest frequencies of STM-specific cytokine-producing CD4+ T cells were found in lamina propria lymphocytes of jejunum and ileum. Significant differences of the relative abundance of cytokine-producing phenotypes between control group and vaccinated + infected animals were detected in most organs, but dominated in gut and lymph node-residing CD4+ T cells. IL-17A producing CD4+ T cells dominated in gut and gut-draining lymph nodes, whereas IFN-γ/TNF-α co-producing CD4+ T cells were present in all locations. Additionally, the majority of cytokine-producing CD4+ T cells had a CD8α+CD27- phenotype, indicative of a late effector or effector memory stage of differentiation. In summary, we show that Salmonella-specific multifunctional CD4+ T cells exist in vaccinated and infected pigs, dominate in the gut and most likely contribute to protective immunity against STM in the pig.
Collapse
Affiliation(s)
- Selma Schmidt
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Elena L Sassu
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eleni Vatzia
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Alix Pierron
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Julia Lagler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.,Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Kerstin H Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Christian Knecht
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Joachim Spergser
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | | | - Tobias Theuß
- Ceva Innovation Center GmbH, Dessau-Roßlau, Germany
| | | | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
32
|
Jin C, Hill J, Gunn BM, Yu WH, Dahora LC, Jones E, Johnson M, Gibani MM, Spreng RL, Alam SM, Nebykova A, Juel HB, Dennison SM, Seaton KE, Fallon JK, Tomaras GD, Alter G, Pollard AJ. Vi-specific serological correlates of protection for typhoid fever. J Exp Med 2020; 218:211531. [PMID: 33180929 PMCID: PMC7668386 DOI: 10.1084/jem.20201116] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/13/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Typhoid Vi vaccines have been shown to be efficacious in children living in endemic regions; however, a widely accepted correlate of protection remains to be established. We applied a systems serology approach to identify Vi-specific serological correlates of protection using samples obtained from participants enrolled in an experimental controlled human infection study. Participants were vaccinated with Vi-tetanus toxoid conjugate (Vi-TT) or unconjugated Vi-polysaccharide (Vi-PS) vaccines and were subsequently challenged with Salmonella Typhi bacteria. Multivariate analyses identified distinct protective signatures for Vi-TT and Vi-PS vaccines in addition to shared features that predicted protection across both groups. Vi IgA quantity and avidity correlated with protection from S. Typhi infection, whereas higher fold increases in Vi IgG responses were associated with reduced disease severity. Targeted antibody-mediated functional responses, particularly neutrophil phagocytosis, were also identified as important components of the protective signature. These humoral markers could be used to evaluate and develop efficacious Vi-conjugate vaccines and assist with accelerating vaccine availability to typhoid-endemic regions.
Collapse
Affiliation(s)
- Celina Jin
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Bronwyn M Gunn
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Wen-Han Yu
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Lindsay C Dahora
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Mari Johnson
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Malick M Gibani
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Rachel L Spreng
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - S Munir Alam
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Anna Nebykova
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - Helene B Juel
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| | - S Moses Dennison
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Kelly E Seaton
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Jonathan K Fallon
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Georgia D Tomaras
- Departments of Immunology, Surgery, and Molecular Genetics and Microbiology, Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
33
|
Stévenin V, Chang YY, Le Toquin Y, Duchateau M, Gianetto QG, Luk CH, Salles A, Sohst V, Matondo M, Reiling N, Enninga J. Dynamic Growth and Shrinkage of the Salmonella-Containing Vacuole Determines the Intracellular Pathogen Niche. Cell Rep 2020; 29:3958-3973.e7. [PMID: 31851926 PMCID: PMC6931108 DOI: 10.1016/j.celrep.2019.11.049] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/23/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
Salmonella is a human and animal pathogen that causes gastro-enteric diseases. The key to Salmonella infection is its entry into intestinal epithelial cells, where the bacterium resides within a Salmonella-containing vacuole (SCV). Salmonella entry also induces the formation of empty macropinosomes, distinct from the SCV, in the vicinity of the entering bacteria. A few minutes after its formation, the SCV increases in size through fusions with the surrounding macropinosomes. Salmonella also induces membrane tubules that emanate from the SCV and lead to SCV shrinkage. Here, we show that these antipodal events are utilized by Salmonella to either establish a vacuolar niche or to be released into the cytosol by SCV rupture. We identify the molecular machinery underlying dynamic SCV growth and shrinkage. In particular, the SNARE proteins SNAP25 and STX4 participate in SCV inflation by fusion with macropinosomes. Thus, host compartment size control emerges as a pathogen strategy for intracellular niche regulation. The early SCV simultaneously grows and shrinks through fusion and tubule formation SCV shrinkage promotes vacuolar rupture and cytosolic release IAMs are enriched in the host SNAREs SNAP25 and STX4, enabling IAM-SCV fusion Promoting SNX1-mediated tubule formation, SopB fosters SCV ruptures
Collapse
Affiliation(s)
- Virginie Stévenin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, Paris, France
| | - Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, Paris, France
| | - Yoann Le Toquin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, Paris, France
| | - Magalie Duchateau
- Institut Pasteur, Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, C2RT, USR 2000 CNRS, Paris, France
| | - Quentin Giai Gianetto
- Institut Pasteur, Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, C2RT, USR 2000 CNRS, Paris, France; Institut Pasteur, Bioinformatics and Biostatistics HUB, C3BI, USR CNRS 3756, Paris, France
| | - Chak Hon Luk
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, Paris, France
| | - Audrey Salles
- Institut Pasteur, UtechS Photonic BioImaging PBI (Imagopole), Centre de Recherche et de Ressources Technologiques C2RT, Paris, France
| | - Victoria Sohst
- Research Center Borstel, Leibniz Lung Center, RG Microbial Interface Biology, Parkallee 22, 23845 Borstel, Germany
| | - Mariette Matondo
- Institut Pasteur, Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, C2RT, USR 2000 CNRS, Paris, France
| | - Norbert Reiling
- Research Center Borstel, Leibniz Lung Center, RG Microbial Interface Biology, Parkallee 22, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, Paris, France.
| |
Collapse
|
34
|
Luo W, Qin H, Chen D, Wu M, Meng K, Zhang A, Pan Y, Qu W, Xie S. The dose regimen formulation of tilmicosin against Lawsonia intracellularis in pigs by pharmacokinetic-pharmacodynamic (PK-PD) model. Microb Pathog 2020; 147:104389. [PMID: 32707311 DOI: 10.1016/j.micpath.2020.104389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 11/25/2022]
Abstract
In this study, the rational dose regimens of tilmicosin against Lawsonia intracellularis (L. intracellularis) were studied using pharmacokinetic-pharmacodynamic (PK-PD) model approach to provide a maximal efficacy. The healthy and infected pigs were orally administrated the tilmicosin premix at a single dose of 10 mg/kg, and then the plasma and ileum content were collected at different time points. The time to peak (Tmax), the peak concentration (Cmax), the area under concentration time curve (AUC0-24h), the apparent volume of distribution by bioavailability (V/F), the body clearance rate by bioavailability (CL/F) and the mean residence time (MRT) of tilmicosin premix for plasma were 2.00 h, 1.08 ± 0.04 μg/mL, 9.61 ± 1.47 μg h/mL, 34.43 ± 1.02 L/kg, 0.71 ± 0.03 L/h/kg and 15.03 ± 0.04 h in healthy pigs, and 2.00 h, 0.99 ± 0.03 μg/mL, 9.30 ± 1.43 μg h/mL, 58.59 ± 1.81 L/kg, 0.44 ± 0.02 L/h/kg and 15.75 ± 0.03 h in infected pigs, respectively. The Tmax, Cmax, AUC0-24h, V/F, CL/F and MRT of tilmicosin premix for ileum content were 2.00 h, 3.78 ± 0.03 μg/mL, 20.41 ± 1.64 μg h/mL, 11.29 ± 0.97 L/kg, 0.44 ± 0.02 L/h/kg and 11.29 ± 0.09 h in healthy pigs, and 2.00 h, 3.41 ± 0.06 μg/mL, 22.65 ± 1.32 μg h/mL, 8.16 ± 1.51 L/kg, 0.41 ± 0.01 L/h/kg and 11.44 ± 0.05 h in infected pigs, respectively. Based on the intracellular minimum inhibitory concentration (MIC) of L. intracellularis isolate was 2 μg/mL, the results of the mutant prevention concentration (MPC), the post-antibiotic effect (PAE) and time-killing curves all showed strong concentration-dependenttendencies. Integrating the in vivo pharmacokinetic data of infected pigs and ex vivo pharmacodynamic data using the sigmoid Emax (Hill) equation to obtain the ileum content AUC0-24h/MIC values of 6.87, 26.80, and 36.02 h to achieve the bacteriostatic activity, bactericidal activity, and virtual eradication of bacteria, respectively. Based on these results, a dosage regimen of daily 14.39 mg/kg for 3 d could be sufficient in the treatment of L. intracellularis. This study will provide a guidance of dosage regimen formulation for drug against animal intracellular bacterial infections.
Collapse
Affiliation(s)
- Wanhe Luo
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Hua Qin
- Beijing TEAM Junwei Healthcare Technology Development Co., Ltd., Beijing, 102600, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China; MARA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Mengru Wu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Kuiyu Meng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Aoxue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Yunahu Pan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Wei Qu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
35
|
Tsai CN, MacNair CR, Cao MPT, Perry JN, Magolan J, Brown ED, Coombes BK. Targeting Two-Component Systems Uncovers a Small-Molecule Inhibitor of Salmonella Virulence. Cell Chem Biol 2020; 27:793-805.e7. [PMID: 32413287 DOI: 10.1016/j.chembiol.2020.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/27/2022]
Abstract
Salmonella serovars are leading causes of gastrointestinal disease and have become increasingly resistant to fluoroquinolone and cephalosporin antibiotics. Overcoming this healthcare crisis requires new approaches in antibiotic discovery and the identification of unique bacterial targets. In this work, we describe a chemical genomics approach to identify inhibitors of Salmonella virulence. From a cell-based, promoter reporter screen of ∼50,000 small molecules, we identified dephostatin as a non-antibiotic compound that inhibits intracellular virulence factors and polymyxin resistance genes. Dephostatin disrupts signaling through both the SsrA-SsrB and PmrB-PmrA two-component regulatory systems and restores sensitivity to the last-resort antibiotic, colistin. Cell-based experiments and mouse models of infection demonstrate that dephostatin attenuates Salmonella virulence in vitro and in vivo, suggesting that perturbing regulatory networks is a promising strategy for the development of anti-infectives.
Collapse
Affiliation(s)
- Caressa N Tsai
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Craig R MacNair
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - My P T Cao
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jordyn N Perry
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Brian K Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
36
|
Verma S, Prescott RA, Ingano L, Nickerson KP, Hill E, Faherty CS, Fasano A, Senger S, Cherayil BJ. The YrbE phospholipid transporter of Salmonella enterica serovar Typhi regulates the expression of flagellin and influences motility, adhesion and induction of epithelial inflammatory responses. Gut Microbes 2020; 11:526-538. [PMID: 31829769 PMCID: PMC7527071 DOI: 10.1080/19490976.2019.1697593] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
serovar Typhi is the etiologic agent of typhoid fever, a major public health problem in the developing world. Moving toward and adhering to the intestinal epithelium represents key initial steps of infection by S. Typhi. We examined the role of the S. Typhi yrbE gene, which encodes an inner membrane phospholipid transporter, in these interactions with epithelial cells. Disruption of yrbE resulted in elevated expression of flagellin and a hypermotile phenotype. It also significantly reduced the ability of S. Typhi to adhere to the HeLa epithelial cell line and to polarized primary epithelial cells derived from human ileal organoids. Interestingly, the yrbE-deficient strain of S. Typhi induced higher production of interleukin-8 from the primary human ileal epithelial cell monolayers compared to the wild-type bacteria. Deletion of the flagellin gene (fliC) in the yrbE-deficient S. Typhi inhibited motility and attenuated interleukin-8 production, but it did not correct the defect in adhesion. We also disrupted yrbE in S. Typhimurium. In contrast to the results in S. Typhi, the deficiency of yrbE in S. Typhimurium had no significant effect on flagellin expression, motility or adhesion to HeLa cells. Correspondingly, the lack of yrbE also had no effect on association with the intestine or the severity of intestinal inflammation in the mouse model of S. Typhimurium infection. Thus, our results point to an important and serovar-specific role played by yrbE in the early stages of intestinal infection by S. Typhi.
Collapse
Affiliation(s)
- Smriti Verma
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel A. Prescott
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laura Ingano
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kourtney P. Nickerson
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Emily Hill
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, USA,CONTACT Bobby J. Cherayil Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
37
|
A Genome-Wide Knockout Screen in Human Macrophages Identified Host Factors Modulating Salmonella Infection. mBio 2019; 10:mBio.02169-19. [PMID: 31594818 PMCID: PMC6786873 DOI: 10.1128/mbio.02169-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A genome-scale CRISPR knockout library screen of THP-1 human macrophages was performed to identify loss-of-function mutations conferring resistance to Salmonella uptake. The screen identified 183 candidate genes, from which 14 representative genes involved in actin dynamics (ACTR3, ARPC4, CAPZB, TOR3A, CYFIP2, CTTN, and NHLRC2), glycosaminoglycan metabolism (B3GNT1), receptor signaling (PDGFB and CD27), lipid raft formation (CLTCL1), calcium transport (ATP2A2 and ITPR3), and cholesterol metabolism (HMGCR) were analyzed further. For some of these pathways, known chemical inhibitors could replicate the Salmonella resistance phenotype, indicating their potential as targets for host-directed therapy. The screen indicated a role for the relatively uncharacterized gene NHLRC2 in both Salmonella invasion and macrophage differentiation. Upon differentiation, NHLRC2 mutant macrophages were hyperinflammatory and did not exhibit characteristics typical of macrophages, including atypical morphology and inability to interact and phagocytose bacteria/particles. Immunoprecipitation confirmed an interaction of NHLRC2 with FRYL, EIF2AK2, and KLHL13.IMPORTANCE Salmonella exploits macrophages to gain access to the lymphatic system and bloodstream to lead to local and potentially systemic infections. With an increasing number of antibiotic-resistant isolates identified in humans, Salmonella infections have become major threats to public health. Therefore, there is an urgent need to identify alternative approaches to anti-infective therapy, including host-directed therapies. In this study, we used a simple genome-wide screen to identify 183 candidate host factors in macrophages that can confer resistance to Salmonella infection. These factors may be potential therapeutic targets against Salmonella infections.
Collapse
|
38
|
Berkowitz L, Pardo-Roa C, Salazar GA, Salazar-Echegarai F, Miranda JP, Ramírez G, Chávez JL, Kalergis AM, Bueno SM, Álvarez-Lobos M. Mucosal Exposure to Cigarette Components Induces Intestinal Inflammation and Alters Antimicrobial Response in Mice. Front Immunol 2019; 10:2289. [PMID: 31608070 PMCID: PMC6773925 DOI: 10.3389/fimmu.2019.02289] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022] Open
Abstract
The main environmental risk factor associated with the development of Crohn's disease (CD) is cigarette smoking. Although the mechanism is still unknown, some studies have shown that cigarette exposure affects the intestinal barrier of the small bowel. Among the factors that may be involved in this process are Paneth cells. These specialized epithelial cells are located into the small intestine, and they are able to secrete antimicrobial peptides, having an essential role in the control of the growth of microorganisms. Alterations in its function are associated with inflammatory processes, such as CD. To study how cigarette components impact ileum homeostasis and Paneth cells integrity, we used intragastric administration of cigarette smoke condensate (CSC) in mice. Our results showed that inflammation was triggered after mucosal exposure of CSC, which induced particular alterations in Paneth cells granules, antimicrobial peptide production, and a reduction of bactericidal capacity. In fact, exposure to CSC generated an imbalance in the fecal bacterial population and increased the susceptibility of mice to develop ileal damage in response to bacterial infection. Moreover, our results obtained in mice unable to produce interleukin 10 (IL-10−/− mice) suggest that CSC treatment can induce a symptomatic enterocolitis with a pathological inflammation in genetically susceptible individuals.
Collapse
Affiliation(s)
- Loni Berkowitz
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Pardo-Roa
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Geraldyne A Salazar
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Salazar-Echegarai
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José P Miranda
- Departamento de Nutrición, Diabetes y Metabolismo, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gigliola Ramírez
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José L Chávez
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Álvarez-Lobos
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
39
|
Awofisayo-Okuyelu A, Pratt A, McCarthy N, Hall I. Within-host mathematical modelling of the incubation period of Salmonella Typhi. ROYAL SOCIETY OPEN SCIENCE 2019; 6:182143. [PMID: 31598273 PMCID: PMC6774937 DOI: 10.1098/rsos.182143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 08/06/2019] [Indexed: 06/10/2023]
Abstract
Mechanistic mathematical models are often employed to understand the dynamics of infectious diseases within a population or within a host. They provide estimates that may not be otherwise available. We have developed a within-host mathematical model in order to understand how the pathophysiology of Salmonella Typhi contributes to its incubation period. The model describes the process of infection from ingestion to the onset of clinical illness using a set of ordinary differential equations. The model was parametrized using estimated values from human and mouse experimental studies and the incubation period was estimated as 9.6 days. A sensitivity analysis was also conducted to identify the parameters that most affect the derived incubation period. The migration of bacteria to the caecal lymph node was observed as a major bottle neck for infection. The sensitivity analysis indicated the growth rate of bacteria in late phase systemic infection and the net population of bacteria in the colon as parameters that most influence the incubation period. We have shown in this study how mathematical models aid in the understanding of biological processes and can be used in estimating parameters of infectious diseases.
Collapse
Affiliation(s)
- Adedoyin Awofisayo-Okuyelu
- National Institute of Health Research Health Protection Research Unit in Gastrointestinal Infections, University of Oxford, Oxford, UK
- Department of Zoology, University of Oxford, Oxford, UK
| | - Adrian Pratt
- Emergency Response Department Science and Technology (ERD S&T), Health Protection Directorate, Public Health England, Porton Down, UK
| | - Noel McCarthy
- National Institute of Health Research Health Protection Research Unit in Gastrointestinal Infections, University of Oxford, Oxford, UK
- Department of Zoology, University of Oxford, Oxford, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Ian Hall
- School of Mathematics, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Masud S, van der Burg L, Storm L, Prajsnar TK, Meijer AH. Rubicon-Dependent Lc3 Recruitment to Salmonella-Containing Phagosomes Is a Host Defense Mechanism Triggered Independently From Major Bacterial Virulence Factors. Front Cell Infect Microbiol 2019; 9:279. [PMID: 31428591 PMCID: PMC6688089 DOI: 10.3389/fcimb.2019.00279] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022] Open
Abstract
Intracellular pathogens such as Salmonella depend on their molecular virulence factors to evade host defense responses like autophagy. Using a zebrafish systemic infection model, we have previously shown that phagocytes, predominantly macrophages, target Salmonella Typhimurium by an autophagy-related pathway known as Lc3-associated phagocytosis (LAP), which is dependent on the host protein Rubicon. Here, we explore the influence of Salmonella virulence factors on pathogenicity in the zebrafish model and induction of LAP as a defense response. We investigated five mutant strains that all could trigger GFP-Lc3 recruitment as puncta or rings around single bacteria or bacterial clusters, in a Rubicon-dependent manner. We found that S. Typhimurium strains carrying mutations in PhoP or PurA, responsible for adaptation to the intracellular environment and efficient metabolism of purines, respectively, are attenuated in the zebrafish model. However, both strains show increased virulence when LAP is inhibited by knockdown of Rubicon. Mutations in type III secretion systems 1 and 2, SipB and SsrB, which are important for invading and replicating in non-phagocytic cells, did not affect the ability to establish successful infection in the zebrafish model. This observation is in line with our previous characterization of this infection model revealing that macrophages actively phagocytose the majority of S. Typhimurium. In contrast to SipB mutants, SsrB mutants were unable to become more virulent in Rubicon-deficient hosts, suggesting that type III system 2 effectors are important for intracellular replication of Salmonella in the absence of LAP. Finally, we found that mutation of FlhD, required for production of flagella, renders S. Typhimurium hypervirulent both in wild type zebrafish embryos and in Rubicon-deficient hosts. FlhD mutation also led to lower levels of GFP-Lc3 recruitment compared with the wild type strain, indicating that recognition of flagellin by the host innate immune system promotes the LAP response. Together, our results provide new evidence that the Rubicon-dependent LAP process is an important defense mechanism against S. Typhimurium.
Collapse
Affiliation(s)
- Samrah Masud
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | | - Lisanne Storm
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | | | |
Collapse
|
41
|
Ye C, Li Q, Li X, Park CG, He Y, Zhang Y, Wu B, Xue Y, Yang K, Lv Y, Ying XL, Ding HH, Cai H, Alkraiem AA, Njiri O, Tembo J, Huang HP, Li AY, Gong J, Qin J, Cheng B, Wei X, Sun Z, Zhang SS, Zhang P, Zheng GX, Li W, Kan B, Yan M, Xiding X, Huo X, Zeng Y, Peng H, Fu Y, Klena JD, Skurnik M, Jiang LY, Chen T. Salmonella enterica Serovar Typhimurium Interacts with CD209 Receptors To Promote Host Dissemination and Infection. Infect Immun 2019; 87:e00100-19. [PMID: 31085704 PMCID: PMC6652768 DOI: 10.1128/iai.00100-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/04/2019] [Indexed: 12/22/2022] Open
Abstract
Salmonella enterica serovar Typhimurium, a Gram-negative bacterium, can cause infectious diseases ranging from gastroenteritis to systemic dissemination and infection. However, the molecular mechanisms underlying this bacterial dissemination have yet to be elucidated. A study indicated that using the lipopolysaccharide (LPS) core as a ligand, S Typhimurium was able to bind human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (hCD209a), an HIV receptor that promotes viral dissemination by hijacking antigen-presenting cells (APCs). In this study, we showed that S Typhimurium interacted with CD209s, leading to the invasion of APCs and potentially the dissemination to regional lymph nodes, spleen, and liver in mice. Shielding of the exposed LPS core through the expression of O-antigen reduces dissemination and infection. Thus, we propose that similar to HIV, S Typhimurium may also utilize APCs via interactions with CD209s as a way to disseminate to the lymph nodes, spleen, and liver to initiate host infection.
Collapse
Affiliation(s)
- Chenglin Ye
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Qiao Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xinyi Li
- Department of Clinical Laboratory, Jingmen No. 1 People's Hospital, Jingmen, China
| | - Chae Gyu Park
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yingxia He
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Yingmiao Zhang
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Bicong Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Ying Xue
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Kun Yang
- Department of Pathogen Biology and Immunology, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yin Lv
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xiao-Ling Ying
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Hong-Hui Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Huahua Cai
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Ayman Ahmad Alkraiem
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
- Department of Biology, College of Science, Taibah University, Medina, Kingdom of Saudi Arabia
| | - Olivia Njiri
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - John Tembo
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Hong-Ping Huang
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - An-Yi Li
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Jianping Gong
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Jichao Qin
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Bing Cheng
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Ziyong Sun
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Shu-Sheng Zhang
- Department of Biomedical Science, College of Medicine-Rockford, University of Illinois at Chicago, Rockford, Illinois, USA
| | - Pei Zhang
- Department of Biomedical Science, College of Medicine-Rockford, University of Illinois at Chicago, Rockford, Illinois, USA
| | - Guo-Xing Zheng
- Department of Biomedical Science, College of Medicine-Rockford, University of Illinois at Chicago, Rockford, Illinois, USA
| | - Wei Li
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Biao Kan
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Meiying Yan
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Xiamu Xiding
- Division of Disease Control and Prevention for Endemic Diseases, Wenquan Center for Disease Control and Prevention, Xinjiang, China
| | - Xixiang Huo
- Hubei Provincial Center for Disease Control and Prevention (CDC), Wuhan, Hubei, China
| | - Yingchun Zeng
- Hubei Provincial Center for Disease Control and Prevention (CDC), Wuhan, Hubei, China
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yangxin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John D Klena
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Ling-Yu Jiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Tie Chen
- Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| |
Collapse
|
42
|
Hassena AB, Siala M, Guermazi S, Zormati S, Gdoura R, Sellami H. Occurrence and Phenotypic and Molecular Characterization of Antimicrobial Resistance of Salmonella Isolates from Food in Tunisia. J Food Prot 2019; 82:1166-1175. [PMID: 31233356 DOI: 10.4315/0362-028x.jfp-18-607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
HIGHLIGHTS Multidrug-resistant Salmonella isolates have been recovered from food in Tunisia. Salmonella isolates from food are resistant to fluoroquinolones and cephalosporins. Surveillance of the antimicrobial susceptibility of foodborne bacteria is needed in Tunisia.
Collapse
Affiliation(s)
- Amal Ben Hassena
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Mariam Siala
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Sonda Guermazi
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Sonia Zormati
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia.,2 Centre Régional de Recherches Vétérinaires de Sfax, Sfax, Tunisia
| | - Radhouane Gdoura
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Hanen Sellami
- 1 Laboratoire de recherche Toxicologie Microbiologie Environnementale et Santé (LR17ES06), Département des Sciences de la vie, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia.,3 Laboratoire de Traitement et de Valorisation des Rejets Hydriques (LTVRH), Water Researches and Technologies Center (CERTE), University of Carthage, Tourist Road Soliman, BP 273-8020, Nabeul 8000, Tunisia
| |
Collapse
|
43
|
Masud S, Prajsnar TK, Torraca V, Lamers GE, Benning M, Van Der Vaart M, Meijer AH. Macrophages target Salmonella by Lc3-associated phagocytosis in a systemic infection model. Autophagy 2019; 15:796-812. [PMID: 30676840 PMCID: PMC6526873 DOI: 10.1080/15548627.2019.1569297] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 11/08/2022] Open
Abstract
Innate immune defense against intracellular pathogens, like Salmonella, relies heavily on the autophagy machinery of the host. This response is studied intensively in epithelial cells, the target of Salmonella during gastrointestinal infections. However, little is known of the role that autophagy plays in macrophages, the predominant carriers of this pathogen during systemic disease. Here we utilize a zebrafish embryo model to study the interaction of S. enterica serovar Typhimurium with the macroautophagy/autophagy machinery of macrophages in vivo. We show that phagocytosis of live but not heat-killed Salmonella triggers recruitment of the autophagy marker GFP-Lc3 in a variety of patterns labeling tight or spacious bacteria-containing compartments, also revealed by electron microscopy. Neutrophils display similar GFP-Lc3 associations, but genetic modulation of the neutrophil/macrophage balance and ablation experiments show that macrophages are critical for the defense response. Deficiency of atg5 reduces GFP-Lc3 recruitment and impairs host resistance, in contrast to atg13 deficiency, indicating that Lc3-Salmonella association at this stage is independent of the autophagy preinitiation complex and that macrophages target Salmonella by Lc3-associated phagocytosis (LAP). In agreement, GFP-Lc3 recruitment and host resistance are impaired by deficiency of Rubcn/Rubicon, known as a negative regulator of canonical autophagy and an inducer of LAP. We also found strict dependency on NADPH oxidase, another essential factor for LAP. Both Rubcn and NADPH oxidase are required to activate a Salmonella biosensor for reactive oxygen species inside infected macrophages. These results identify LAP as the major host protective autophagy-related pathway responsible for macrophage defense against Salmonella during systemic infection. Abbreviations: ATG: autophagy related gene; BECN1: Beclin 1; CFU: colony forming units; CYBA/P22PHOX: cytochrome b-245, alpha chain; CYBB/NOX2: cytochrome b-245 beta chain; dpf: days post fertilization; EGFP: enhanced green fluorescent protein; GFP: green fluorescent protein; hfp: hours post fertilization; hpi: hours post infection; IRF8: interferon regulatory factor 8; Lcp1/L-plastin: lymphocyte cytosolic protein 1; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1A/1B-light chain 3; mCherry: red fluorescent protein; mpeg1: macrophage expressed gene 1; mpx: myeloid specific peroxidase; NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase; NCF4/P40PHOX: neutrophil cytosolic factor 4; NTR-mCherry: nitroreductase-mCherry fusion; PTU: phenylthiourea; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; RB1CC1/FIP200: RB-1 inducible coiled coin 1; ROS: reactive oxygen species; RT-PCR: reverse transcriptase polymerase chain reaction; RUBCN/RUBICON: RUN and cysteine rich domain containing BECN1-interacting protein; SCV: Salmonella-containing vacuole; S. Typhimurium/S.T: Salmonella enterica serovar Typhimurium; TEM: transmission electron microscopy; Tg: transgenic; TSA: tyramide signal amplification; ULK1/2: unc-51-like autophagy activating kinase 1/2; UVRAG: UVRAG: UV radiation resistance associated; wt: wild type.
Collapse
Affiliation(s)
- Samrah Masud
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | | | - Vincenzo Torraca
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Gerda E.M. Lamers
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Marianne Benning
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | | | | |
Collapse
|
44
|
Ellis MJ, Tsai CN, Johnson JW, French S, Elhenawy W, Porwollik S, Andrews-Polymenis H, McClelland M, Magolan J, Coombes BK, Brown ED. A macrophage-based screen identifies antibacterial compounds selective for intracellular Salmonella Typhimurium. Nat Commun 2019; 10:197. [PMID: 30643129 PMCID: PMC6331611 DOI: 10.1038/s41467-018-08190-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022] Open
Abstract
Salmonella Typhimurium (S. Tm) establishes systemic infection in susceptible hosts by evading the innate immune response and replicating within host phagocytes. Here, we sought to identify inhibitors of intracellular S. Tm replication by conducting parallel chemical screens against S. Tm growing in macrophage-mimicking media and within macrophages. We identify several compounds that inhibit Salmonella growth in the intracellular environment and in acidic, ion-limited media. We report on the antimicrobial activity of the psychoactive drug metergoline, which is specific against intracellular S. Tm. Screening an S. Tm deletion library in the presence of metergoline reveals hypersensitization of outer membrane mutants to metergoline activity. Metergoline disrupts the proton motive force at the bacterial cytoplasmic membrane and extends animal survival during a systemic S. Tm infection. This work highlights the predictive nature of intracellular screens for in vivo efficacy, and identifies metergoline as a novel antimicrobial active against Salmonella.
Collapse
Affiliation(s)
- Michael J Ellis
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Caressa N Tsai
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Jarrod W Johnson
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Wael Elhenawy
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697-4025, USA
| | - Helene Andrews-Polymenis
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA, 92697-4025, USA
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada
| | - Brian K Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
45
|
Vohra P, Chaudhuri RR, Mayho M, Vrettou C, Chintoan-Uta C, Thomson NR, Hope JC, Hopkins J, Stevens MP. Retrospective application of transposon-directed insertion-site sequencing to investigate niche-specific virulence of Salmonella Typhimurium in cattle. BMC Genomics 2019; 20:20. [PMID: 30621582 PMCID: PMC6325888 DOI: 10.1186/s12864-018-5319-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/27/2018] [Indexed: 01/05/2023] Open
Abstract
Background Salmonella enterica subspecies enterica is an animal and zoonotic pathogen of global importance. Cattle are a significant reservoir of human non-typhoidal salmonellosis and can suffer enteric and systemic disease owing to the ability of Salmonella to survive within the bovine lymphatic system and intestines. Contamination of food can occur due to the incorporation of contaminated peripheral lymph nodes or by direct contamination of carcasses with gut contents. It is essential to understand the mechanisms used by Salmonella to enter and persist within the bovine lymphatic system and how they differ from those required for intestinal colonization to minimize zoonotic infections. Results Transposon-directed insertion site sequencing (TraDIS) was applied to pools of mutants recovered from mesenteric lymph nodes (MLNs) draining the distal ileum of calves after oral inoculation with a library of 8550 random S. Typhimurium mini-Tn5Km2 mutants in pools of 475 mutants per calf. A total of 8315 mutants representing 2852 different genes were detected in MLNs and their in vivo fitness was calculated. Using the same improved algorithm for analysis of transposon-flanking sequences, the identity and phenotype of mutants recovered from the distal ileal mucosa of the same calves was also defined, enabling comparison with previously published data and of mutant phenotypes across the tissues. Phenotypes observed for the majority of mutants were highly significantly correlated in the two tissues. However, 32 genes were identified in which transposon insertions consistently resulted in differential fitness in the ileal wall and MLNs, suggesting niche-specific roles for these genes in pathogenesis. Defined null mutations affecting ptsN and spvC were confirmed to result in tissue-specific phenotypes in calves, thus validating the TraDIS dataset. Conclusions This validation of the role of thousands of Salmonella genes and identification of genes with niche-specific roles in a key target species will inform the design of control strategies for bovine salmonellosis and zoonotic infections, for which efficacious and cross-protective vaccines are currently lacking. Electronic supplementary material The online version of this article (10.1186/s12864-018-5319-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Prerna Vohra
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK.
| | - Roy R Chaudhuri
- Department of Molecular Biology and Biotechnology, University of Sheffield, S10 2TN, Sheffield, UK
| | - Matthew Mayho
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Christina Vrettou
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| | - Cosmin Chintoan-Uta
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| | | | - Jayne C Hope
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| | - John Hopkins
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| | - Mark P Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK
| |
Collapse
|
46
|
He YX, Ye CL, Zhang P, Li Q, Park CG, Yang K, Jiang LY, Lv Y, Ying XL, Ding HH, Huang HP, Mambwe Tembo J, Li AY, Cheng B, Zhang SS, Zheng GX, Chen SY, Li W, Xia LX, Kan B, Wang X, Jing HQ, Yang RF, Peng H, Fu YX, Klena JD, Skurnik M, Chen T. Yersinia pseudotuberculosis Exploits CD209 Receptors for Promoting Host Dissemination and Infection. Infect Immun 2019; 87:e00654-18. [PMID: 30348825 PMCID: PMC6300620 DOI: 10.1128/iai.00654-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
Abstract
Yersinia pseudotuberculosis is a Gram-negative enteropathogen and causes gastrointestinal infections. It disseminates from gut to mesenteric lymph nodes (MLNs), spleen, and liver of infected humans and animals. Although the molecular mechanisms for dissemination and infection are unclear, many Gram-negative enteropathogens presumably invade the small intestine via Peyer's patches to initiate dissemination. In this study, we demonstrate that Y. pseudotuberculosis utilizes its lipopolysaccharide (LPS) core to interact with CD209 receptors, leading to invasion of human dendritic cells (DCs) and murine macrophages. These Y. pseudotuberculosis-CD209 interactions result in bacterial dissemination to MLNs, spleens, and livers of both wild-type and Peyer's patch-deficient mice. The blocking of the Y. pseudotuberculosis-CD209 interactions by expression of O-antigen and with oligosaccharides reduces infectivity. Based on the well-documented studies in which HIV-CD209 interaction leads to viral dissemination, we therefore propose an infection route for Y. pseudotuberculosis where this pathogen, after penetrating the intestinal mucosal membrane, hijacks the Y. pseudotuberculosis-CD209 interaction antigen-presenting cells to reach their target destinations, MLNs, spleens, and livers.
Collapse
Affiliation(s)
- Ying-Xia He
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Cheng-Lin Ye
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Pei Zhang
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Qiao Li
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Chae Gyu Park
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kun Yang
- Department of Pathogen Biology and Immunology, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Ling-Yu Jiang
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Yin Lv
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xiao-Ling Ying
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Hong-Hui Ding
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Hong-Ping Huang
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - John Mambwe Tembo
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
- Department of Paediatrics & Child Health, The University of Zambia-University College London Medical School at Zambia, Lusaka, Zambia
| | - An-Yi Li
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Bing Cheng
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Shu-Sheng Zhang
- Department of Biomedical Science, College of Medicine-Rockford, University of Illinois at Chicago, Rockford, Illinois, USA
| | - Guo-Xing Zheng
- Department of Biomedical Science, College of Medicine-Rockford, University of Illinois at Chicago, Rockford, Illinois, USA
| | - Shi-Yun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Li
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Lian-Xu Xia
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Biao Kan
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Xin Wang
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Huai-Qi Jing
- Department of Diarrheal Diseases, National Institute for Communicable Diseases Control and Prevention, Beijing, China
| | - Rui-Fu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hua Peng
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John D Klena
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Tie Chen
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| |
Collapse
|
47
|
Bravo-Blas A, Utriainen L, Clay SL, Kästele V, Cerovic V, Cunningham AF, Henderson IR, Wall DM, Milling SWF. Salmonella enterica Serovar Typhimurium Travels to Mesenteric Lymph Nodes Both with Host Cells and Autonomously. THE JOURNAL OF IMMUNOLOGY 2018; 202:260-267. [PMID: 30487173 PMCID: PMC6305795 DOI: 10.4049/jimmunol.1701254] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/29/2018] [Indexed: 12/31/2022]
Abstract
Salmonella infection is a globally important cause of gastroenteritis and systemic disease and is a useful tool to study immune responses in the intestine. Although mechanisms leading to immune responses against Salmonella have been extensively studied, questions remain about how bacteria travel from the intestinal mucosa to the mesenteric lymph nodes (MLN), a key site for Ag presentation. In this study, we used a mouse model of infection with Salmonella enterica serovar Typhimurium (STM) to identify changes in intestinal immune cells induced during early infection. We then used fluorescently labeled STM to identify interactions with immune cells from the site of infection through migration in lymph to the MLN. We show that viable STM can be carried in the lymph by any subset of migrating dendritic cells but not by macrophages. Moreover, approximately half of the STM in lymph are not associated with cells at all and travel autonomously. Within the MLN, STM associates with dendritic cells and B cells but predominantly with MLN-resident macrophages. In conclusion, we describe the routes used by STM to spread systemically in the period immediately postinfection. This deeper understanding of the infection process could open new avenues for controlling it.
Collapse
Affiliation(s)
- Alberto Bravo-Blas
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom;
| | - Lotta Utriainen
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Slater L Clay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Verena Kästele
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Vuk Cerovic
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom; and
| | - Ian R Henderson
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Daniel M Wall
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Simon W F Milling
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
48
|
Wang H, Owen DL, Qian LJ, Chopp LB, Farrar MA, Hogquist KA. High-throughput Microscopic Analysis of Salmonella Invasion of Host Cells. Bio Protoc 2018; 8:e3017. [PMID: 30345327 PMCID: PMC6195197 DOI: 10.21769/bioprotoc.3017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/06/2018] [Accepted: 08/23/2018] [Indexed: 11/02/2022] Open
Abstract
Salmonella is a Gram-negative bacterium causing a gastro-enteric disease called salmonellosis. During the first phase of infection, Salmonella uses its flagella to swim near the surface of the epithelial cells and to target specific site of infection. In order to study the selection criteria that determine which host cells are targeted by the pathogen, and to analyze the relation between infecting Salmonella (i.e., cooperation or competition), we have established a high-throughput microscopic assay of HeLa cells sequentially infected with fluorescent bacteria. Using an automated pipeline of image analysis, we quantitatively characterized a multitude of parameters of infected and non-infected cells. Based on this, we established a predictive model that allowed us to identify those parameters involved in host cell vulnerability towards infection. We revealed that host cell vulnerability has two origins: a pathogen-induced cellular vulnerability emerging from Salmonella uptake and persisting at later stages of the infection process; and a host cell-inherent vulnerability linked with cell inherent attributes, such as local cell crowding, and cholesterol content. Our method forecasts the probability of Salmonella infection within monolayers of epithelial cells based on morphological or molecular host cell parameters. Here, we provide a detailed description of the workflow including the computer-based analysis pipeline. Our method has the potential to be applied to study other combinations of host-pathogen interactions.
Collapse
Affiliation(s)
- Haiguang Wang
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David L. Owen
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Lily J. Qian
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Laura B. Chopp
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Michael A. Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
49
|
Hefele M, Stolzer I, Ruder B, He GW, Mahapatro M, Wirtz S, Neurath MF, Günther C. Intestinal epithelial Caspase-8 signaling is essential to prevent necroptosis during Salmonella Typhimurium induced enteritis. Mucosal Immunol 2018; 11:1191-1202. [PMID: 29520026 DOI: 10.1038/s41385-018-0011-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 01/03/2018] [Accepted: 01/14/2018] [Indexed: 02/04/2023]
Abstract
Although induction of host cell death is a pivotal step during bacteria-induced gastroenteritis, the molecular regulation remains to be fully characterized. To expand our knowledge, we investigated the role of the central cell death regulator Caspase-8 in response to Salmonella Typhimurium. Here, we uncovered that intestinal salmonellosis was associated with strong upregulation of members of the host cell death machinery in intestinal epithelial cells (IECs) as an early event, suggesting that elimination of infected IECs represents a host defense strategy. Indeed, Casp8∆IEC mice displayed severe tissue damage and high lethality after infection. Additional deletion of Ripk3 or Mlkl rescued epithelial cell death and lethality of Casp8∆IEC mice, demonstrating the crucial role of Caspase-8 as a negative regulator of necroptosis. While Casp8∆IECTnfr1-/- mice showed improved survival after infection, tissue destruction was similar to Casp8∆IEC mice, indicating that necroptosis partially depends on TNF-α signaling. Although there was no impairment in antimicrobial peptide secretion during the early phase of infection, functional Caspase-8 seems to be required to control pathogen colonization. Collectively, these results demonstrate that Caspase-8 is essential to prevent Salmonella Typhimurium induced enteritis and to ensure host survival by two different mechanisms: maintenance of intestinal barrier function and restriction of pathogen colonization.
Collapse
Affiliation(s)
- Manuela Hefele
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Barbara Ruder
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Gui-Wei He
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
50
|
Identification of Parameters of Host Cell Vulnerability during Salmonella Infection by Quantitative Image Analysis and Modeling. Infect Immun 2017; 86:IAI.00644-17. [PMID: 29084895 DOI: 10.1128/iai.00644-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Salmonella targets and enters epithelial cells at permissive entry sites: some cells are more likely to be infected than others. However, the parameters that lead to host cell heterogeneity are not known. Here, we quantitatively characterized host cell vulnerability to Salmonella infection based on imaged parameters. We performed successive infections of the same host cell population followed by automated high-throughput microscopy and observed that infected cells have a higher probability of being reinfected. Establishing a predictive model, we identified two combined origins of host cell vulnerability: pathogen-induced cellular vulnerability emerging from Salmonella uptake and persisting at later stages of the infection and host cell-inherent vulnerability. We linked the host cell-inherent vulnerability with its morphological attributes, such as local cell crowding, and with host cell cholesterol content. This showed that the probability of Salmonella infection success can be forecast from morphological or molecular host cell parameters.
Collapse
|