1
|
Grote A, Hendin N, Amit S, Adani B, Rahav G, Adler A, Livny J, Gal-Mor O, Earl AM. Genetic diversity of Salmonella enterica during acute human infections. Gut Microbes 2025; 17:2491666. [PMID: 40260673 PMCID: PMC12026202 DOI: 10.1080/19490976.2025.2491666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/27/2025] [Accepted: 04/05/2025] [Indexed: 04/24/2025] Open
Abstract
The ubiquitous bacterial pathogen Salmonella enterica is the causative agent of both enteric fever and gastroenteritis. Despite its significant global health burden, we lack an understanding of its genetic diversity during acute infection, with ramifications for treatment and prevention. Here, we investigated within-host infection diversity of acute salmonellosis using whole-genome sequencing of blood or stool isolates obtained from 23 different patients. We found that intestinal infections exhibited greater genetic variation than blood infections, including in their plasmid content. While same-patient isolates were separated by 10 single nucleotide polymorphisms or less, they often differed in the carriage of genes or alleles, including those associated with antibiotic resistance or virulence. Given the longstanding emphasis on single colony isolation in clinical and laboratory microbiology, these findings have implications for how we both study evolution and transmission and how we treat salmonellosis in an age of increasing antibiotic resistance.
Collapse
Affiliation(s)
- Alexandra Grote
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Natav Hendin
- The Infectious Diseases unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Sharon Amit
- Microbiology Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Boaz Adani
- The Infectious Diseases unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Galia Rahav
- The Infectious Diseases unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amos Adler
- Clinical Microbiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Jonathan Livny
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ohad Gal-Mor
- Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Clinical Microbiology and Immunology, Tel-Aviv University, Tel-Aviv, Israel
| | - Ashlee M. Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
2
|
Giorgio RT, Helaine S. Antibiotic-recalcitrant Salmonella during infection. Nat Rev Microbiol 2025; 23:276-287. [PMID: 39558126 DOI: 10.1038/s41579-024-01124-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
3
|
Fruet C, Müller EL, Loverdo C, Bitbol AF. Spatial structure facilitates evolutionary rescue by drug resistance. PLoS Comput Biol 2025; 21:e1012861. [PMID: 40179127 PMCID: PMC11967957 DOI: 10.1371/journal.pcbi.1012861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/09/2025] [Indexed: 04/05/2025] Open
Abstract
Bacterial populations often have complex spatial structures, which can impact their evolution. Here, we study how spatial structure affects the evolution of antibiotic resistance in a bacterial population. We consider a minimal model of spatially structured populations where all demes (i.e., subpopulations) are identical and connected to each other by identical migration rates. We show that spatial structure can facilitate the survival of a bacterial population to antibiotic treatment, starting from a sensitive inoculum. Specifically, the bacterial population can be rescued if antibiotic resistant mutants appear and are present when drug is added, and spatial structure can impact the fate of these mutants and the probability that they are present. Indeed, the probability of fixation of neutral or deleterious mutations providing drug resistance is increased in smaller populations. This promotes local fixation of resistant mutants in the structured population, which facilitates evolutionary rescue by drug resistance in the rare mutation regime. Once the population is rescued by resistance, migrations allow resistant mutants to spread in all demes. Our main result that spatial structure facilitates evolutionary rescue by antibiotic resistance extends to more complex spatial structures, and to the case where there are resistant mutants in the inoculum.
Collapse
Affiliation(s)
- Cecilia Fruet
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Ella Linxia Müller
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS,Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris,France
| | - Anne-Florence Bitbol
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
4
|
Shealy NG, Baltagulov M, de Brito C, McGovern A, Castro P, Schrimpe-Rutledge AC, Malekshahi C, Condreanu SG, Sherrod SD, Jana S, Jones K, Ribeiro TM, McLean JA, Beiting DP, Byndloss MX. Short-term alterations in dietary amino acids override host genetic susceptibility and reveal mechanisms of Salmonella Typhimurium small intestine colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645332. [PMID: 40196486 PMCID: PMC11974825 DOI: 10.1101/2025.03.25.645332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
In addition to individual genetics, environmental factors (e.g., dietary changes) may influence host susceptibility to gastrointestinal infection through unknown mechanisms. Herein, we developed a model in which CBA/J mice, a genetically resistant strain that tolerates intestinal colonization by the enteric pathogen Salmonella Typhimurium (S. Tm), rapidly succumb to infection after exposure to a diet rich in L-amino acids (AA). In mice, S. Tm-gastroenteritis is restricted to the large intestine (cecum), limiting their use to understand S. Tm small intestine (ileum) colonization, a feature of human Salmonellosis. Surprisingly, CBA mice fed AA diet developed ileitis with enhanced S. Tm ileal colonization. Using germ-free mice and ileal-fecal slurry transplant, we found diet-mediated S. Tm ileal expansion to be microbiota-dependent. Mechanistically, S. Tm relied on Fructosyl-asparagine utilization to expand in the ileum during infection. We demonstrate how AA diet overrides host genetics by altering the gut microbiota's ability to prevent S. Tm ileal colonization.
Collapse
Affiliation(s)
- Nicolas G. Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Madi Baltagulov
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Camila de Brito
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Anna McGovern
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Pollyana Castro
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | | | - Clara Malekshahi
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, U. S. A
| | - Simona G. Condreanu
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Stacy D. Sherrod
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Somnath Jana
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Katerina Jones
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - Tamara Machado Ribeiro
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
| | - John A. McLean
- Center for Innovative Technology and Department of Chemistry, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Daniel P. Beiting
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37203, U. S. A
| | - Mariana X. Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37203, U. S. A
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, TN 37235, U.S.A
| |
Collapse
|
5
|
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
|
6
|
Avraham R. Untangling Cellular Host-Pathogen Encounters at Infection Bottlenecks. Infect Immun 2023; 91:e0043822. [PMID: 36939328 PMCID: PMC10112260 DOI: 10.1128/iai.00438-22] [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] [Indexed: 03/21/2023] Open
Abstract
Bacterial pathogens can invade the tissue and establish a protected intracellular niche at the site of invasion that can spread locally (e.g., microcolonies) or to systemic sites (e.g., granulomas). Invasion of the tissue and establishment of intracellular infection are rare events that are difficult to study in the in vivo setting but have critical clinical consequences, such as long-term carriage, reinfections, and emergence of antibiotic resistance. Here, I discuss Salmonella interactions with its host macrophage during early stages of infection and their critical role in determining infection outcome. The dynamics of host-pathogen interactions entail highly heterogenous host immunity, bacterial virulence, and metabolic cross talk, requiring in vivo analysis at single-cell resolution. I discuss models and single-cell approaches that provide a global understanding of the establishment of a protected intracellular niche within the tissue and the host-pathogen landscape at infection bottlenecks during early stages of infection. Studying cellular host-pathogen interactions in vivo can improve our knowledge of the trajectory of infection between the initial inoculation with a dose of pathogens and the appearance of symptoms of disease.
Collapse
Affiliation(s)
- Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
7
|
Hoces D, Greter G, Arnoldini M, Stäubli ML, Moresi C, Sintsova A, Berent S, Kolinko I, Bansept F, Woller A, Häfliger J, Martens E, Hardt WD, Sunagawa S, Loverdo C, Slack E. Fitness advantage of Bacteroides thetaiotaomicron capsular polysaccharide in the mouse gut depends on the resident microbiota. eLife 2023; 12:81212. [PMID: 36757366 PMCID: PMC10014078 DOI: 10.7554/elife.81212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 02/08/2023] [Indexed: 02/10/2023] Open
Abstract
Many microbiota-based therapeutics rely on our ability to introduce a microbe of choice into an already-colonized intestine. In this study, we used genetically barcoded Bacteroides thetaiotaomicron (B. theta) strains to quantify population bottlenecks experienced by a B. theta population during colonization of the mouse gut. As expected, this reveals an inverse relationship between microbiota complexity and the probability that an individual wildtype B. theta clone will colonize the gut. The polysaccharide capsule of B. theta is important for resistance against attacks from other bacteria, phage, and the host immune system, and correspondingly acapsular B. theta loses in competitive colonization against the wildtype strain. Surprisingly, the acapsular strain did not show a colonization defect in mice with a low-complexity microbiota, as we found that acapsular strains have an indistinguishable colonization probability to the wildtype strain on single-strain colonization. This discrepancy could be resolved by tracking in vivo growth dynamics of both strains: acapsular B.theta shows a longer lag phase in the gut lumen as well as a slightly slower net growth rate. Therefore, as long as there is no niche competitor for the acapsular strain, this has only a small influence on colonization probability. However, the presence of a strong niche competitor (i.e., wildtype B. theta, SPF microbiota) rapidly excludes the acapsular strain during competitive colonization. Correspondingly, the acapsular strain shows a similarly low colonization probability in the context of a co-colonization with the wildtype strain or a complete microbiota. In summary, neutral tagging and detailed analysis of bacterial growth kinetics can therefore quantify the mechanisms of colonization resistance in differently-colonized animals.
Collapse
Affiliation(s)
- Daniel Hoces
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Giorgia Greter
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Markus Arnoldini
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Melanie L Stäubli
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Claudia Moresi
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Sara Berent
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Isabel Kolinko
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Florence Bansept
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Aurore Woller
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Janine Häfliger
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Eric Martens
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn ArborUnited States
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| |
Collapse
|
8
|
Wincott CJ, Sritharan G, Benns HJ, May D, Gilabert-Carbajo C, Bunyan M, Fairweather AR, Alves E, Andrew I, Game L, Frickel EM, Tiengwe C, Ewald SE, Child MA. Cellular barcoding of protozoan pathogens reveals the within-host population dynamics of Toxoplasma gondii host colonization. CELL REPORTS METHODS 2022; 2:100274. [PMID: 36046624 PMCID: PMC9421581 DOI: 10.1016/j.crmeth.2022.100274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/20/2022] [Accepted: 07/22/2022] [Indexed: 02/09/2023]
Abstract
Cellular barcoding techniques are powerful tools to understand microbial pathogenesis. However, barcoding strategies have not been broadly applied to protozoan parasites, which have unique genomic structures and virulence strategies compared with viral and bacterial pathogens. Here, we present a CRISPR-based method to barcode protozoa, which we successfully apply to Toxoplasma gondii and Trypanosoma brucei. Using libraries of barcoded T. gondii, we evaluate shifts in the population structure from acute to chronic infection of mice. Contrary to expectation, most barcodes were present in the brain one month post-intraperitoneal infection in both inbred CBA/J and outbred Swiss mice. Although parasite cyst number and barcode diversity declined over time, barcodes representing a minor fraction of the inoculum could become a dominant population in the brain by three months post-infection. These data establish a cellular barcoding approach for protozoa and evidence that the blood-brain barrier is not a major bottleneck to colonization by T. gondii.
Collapse
Affiliation(s)
- Ceire J. Wincott
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Gayathri Sritharan
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
- Department of Biological Sciences, Birkbeck, University of London, Mallet Street, Bloomsbury, London WC1E 7HX, UK
| | - Henry J. Benns
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
- Department of Chemistry, Imperial College London, White City Campus, London W12 0BZ, UK
| | - Dana May
- Department of Microbiology, Immunology and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Carla Gilabert-Carbajo
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Monique Bunyan
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1BF, UK
| | - Aisling R. Fairweather
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Eduardo Alves
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Ivan Andrew
- UKRI London Institute of Medical Sciences Genomics Laboratory, Shepherd’s Bush, London W12 0NN, UK
| | - Laurence Game
- UKRI London Institute of Medical Sciences Genomics Laboratory, Shepherd’s Bush, London W12 0NN, UK
| | - Eva-Maria Frickel
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1BF, UK
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Calvin Tiengwe
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Sarah E. Ewald
- Department of Microbiology, Immunology and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Matthew A. Child
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|
9
|
Transcytosis of IgA Attenuates Salmonella Invasion in Human Enteroids and Intestinal Organoids. Infect Immun 2022; 90:e0004122. [PMID: 35579465 DOI: 10.1128/iai.00041-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Secretory IgA (SIgA) is the most abundant antibody type in intestinal secretions where it contributes to safeguarding the epithelium from invasive pathogens like the Gram-negative bacterium, Salmonella enterica serovar Typhimurium (STm). For example, we recently reported that passive oral administration of the recombinant monoclonal SIgA antibody, Sal4, to mice promotes STm agglutination in the intestinal lumen and restricts bacterial invasion of Peyer's patch tissues. In this report, we sought to recapitulate Sal4-mediated protection against STm in human Enteroids and human intestinal organoids (HIOs) as models to decipher the molecular mechanisms by which antibodies function in mucosal immunity in the human gastrointestinal tract. We confirm that Enteroids and HIO-derived monolayers are permissive to STm infection, dependent on HilD, the master transcriptional regulator of the SPI-I type three secretion system (T3SS). Stimulation of M-like cells in both Enteroids and HIOs by the addition of RANKL further enhanced STm invasion. The apical addition of Sal4 mouse IgA, as well as recombinant human Sal4 dimeric IgA (dIgA) and SIgA resulted a dose-dependent reduction in bacterial invasion. Moreover, basolateral application of Sal4 dIgA to Enteroid and HIO monolayers gave rise to SIgA in the apical compartment via a pathway dependent on expression of the polymeric immunoglobulin receptor (pIgR). The resulting Sal4 SIgA was sufficient to reduce STm invasion of Enteroid and HIO epithelial cell monolayers by ~20-fold. Recombinant Sal4 IgG was also transported in the Enteroid and HIOs, but to a lesser degree and via a pathway dependent on the neonatal Fc receptor (FCGRT). The models described lay the foundation for future studies into detailed mechanisms of IgA and IgG protection against STm and other pathogens.
Collapse
|
10
|
Richards A, Baranova D, Mantis NJ. The prospect of orally administered monoclonal secretory IgA (SIgA) antibodies to prevent enteric bacterial infections. Hum Vaccin Immunother 2022; 18:1964317. [PMID: 34491878 PMCID: PMC9103515 DOI: 10.1080/21645515.2021.1964317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/08/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eliminating diarrheal diseases as a leading cause of childhood morbidity and mortality in low- and middle-income countries (LMICs) will require multiple intervention strategies. In this review, we spotlight a series of preclinical studies investigating the potential of orally administered monoclonal secretory IgA (SIgA) antibodies (MAbs) to reduce disease associated with three enteric bacterial pathogens: Campylobacter jejuni, enterotoxigenic Escherichia coli (ETEC), and invasive Salmonella enterica serovar Typhimurium. IgA MAbs targeting bacterial surface antigens (flagella, adhesins, and lipopolysaccharide) were generated from mice, humanized mice, and human tonsillar B cells. Recombinant SIgA1 and/or SIgA2 derivates of those MAbs were purified from supernatants following transient transfection of 293 cells with plasmids encoding antibody heavy and light chains, J-chain, and secretory component (SC). When administered to mice by gavage immediately prior to (or admixed with) the bacterial challenge, SIgA MAbs reduced infection C. jejuni, ETEC, and S. Typhimurium infections. Fv-matched IgG1 MAbs by comparison were largely ineffective against C. jejuni and S. Typhimurium under the same conditions, although they were partially effective against ETEC. While these findings highlight future applications of orally administered SIgA, the studies also underscored the fundamental challenges associated with using MAbs as prophylactic tools against enteric bacterial diseases.
Collapse
Affiliation(s)
- Angelene Richards
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Danielle Baranova
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
11
|
Fulde M, van Vorst K, Zhang K, Westermann AJ, Busche T, Huei YC, Welitschanski K, Froh I, Pägelow D, Plendl J, Pfarrer C, Kalinowski J, Vogel J, Valentin-Weigand P, Hensel M, Tedin K, Repnik U, Hornef MW. SPI2 T3SS effectors facilitate enterocyte apical to basolateral transmigration of Salmonella-containing vacuoles in vivo. Gut Microbes 2022; 13:1973836. [PMID: 34542008 PMCID: PMC8475570 DOI: 10.1080/19490976.2021.1973836] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Salmonella pathogenicity island (SPI) 2 type three secretion system (T3SS)-mediated effector molecules facilitate bacterial survival in phagocytes but their role in the intestinal epithelium in vivo remains ill-defined. Using our neonatal murine infection model in combination with SPI2 reporter technology and RNA-Seq of sorted primary enterocytes, we demonstrate expression of SPI2 effector molecules by intraepithelial Salmonella Typhimurium (S. Typhimurium). Contrary to expectation, immunostaining revealed that infection with SPI2 T3SS-mutants resulted in significantly enlarged intraepithelial Salmonella-containing vacuoles (SCV) with altered cellular positioning, suggesting impaired apical to basolateral transmigration. Also, infection with isogenic tagged S. Typhimurium strains revealed a reduced spread of intraepithelial SPI2 T3SS mutant S. Typhimurium to systemic body sites. These results suggest that SPI2 T3SS effector molecules contribute to enterocyte apical to basolateral transmigration of the SCV during the early stage of the infection.
Collapse
Affiliation(s)
- Marcus Fulde
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Microbiology and Epizootics, Berlin, Germany,CONTACT Mathias Hornef Institute for Medical Microbiology; RWTH University Hospital; Pauwelsstr. 30, Aachen, D-52074, Germany
| | - Kira van Vorst
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Microbiology and Epizootics, Berlin, Germany
| | - Kaiyi Zhang
- Institute of Medical Microbiology, Rwth University Hospital Aachen, Aachen, Germany
| | - Alexander J. Westermann
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany,Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Tobias Busche
- Technology Platform Genomics, Center for Biotechnology (Cebitec), Bielefeld University, Bielefeld, Germany
| | - Yong Chiun Huei
- Institute of Medical Microbiology, Rwth University Hospital Aachen, Aachen, Germany
| | - Katharina Welitschanski
- Hannover Medical School, Institute for Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Isabell Froh
- Hannover Medical School, Institute for Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Dennis Pägelow
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Microbiology and Epizootics, Berlin, Germany
| | - Johanna Plendl
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Veterinary Anatomy, Berlin, Germany
| | - Christiane Pfarrer
- Institute for Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Jörn Kalinowski
- Technology Platform Genomics, Center for Biotechnology (Cebitec), Bielefeld University, Bielefeld, Germany
| | - Jörg Vogel
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany,Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Peter Valentin-Weigand
- Institute of Microbiology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Michael Hensel
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Karsten Tedin
- Department of Veterinary Medicine, Freie Universität Berlin, Institute of Microbiology and Epizootics, Berlin, Germany
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Mathias W. Hornef
- Institute of Medical Microbiology, Rwth University Hospital Aachen, Aachen, Germany
| |
Collapse
|
12
|
Siggins MK, Sriskandan S. Bacterial Lymphatic Metastasis in Infection and Immunity. Cells 2021; 11:33. [PMID: 35011595 PMCID: PMC8750085 DOI: 10.3390/cells11010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Lymphatic vessels permeate tissues around the body, returning fluid from interstitial spaces back to the blood after passage through the lymph nodes, which are important sites for adaptive responses to all types of pathogens. Involvement of the lymphatics in the pathogenesis of bacterial infections is not well studied. Despite offering an obvious conduit for pathogen spread, the lymphatic system has long been regarded to bar the onward progression of most bacteria. There is little direct data on live virulent bacteria, instead understanding is largely inferred from studies investigating immune responses to viruses or antigens in lymph nodes. Recently, we have demonstrated that extracellular bacterial lymphatic metastasis of virulent strains of Streptococcus pyogenes drives systemic infection. Accordingly, it is timely to reconsider the role of lymph nodes as absolute barriers to bacterial dissemination in the lymphatics. Here, we summarise the routes and mechanisms by which an increasing variety of bacteria are acknowledged to transit through the lymphatic system, including those that do not necessarily require internalisation by host cells. We discuss the anatomy of the lymphatics and other factors that influence bacterial dissemination, as well as the consequences of underappreciated bacterial lymphatic metastasis on disease and immunity.
Collapse
Affiliation(s)
- Matthew K. Siggins
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
13
|
Mahmutovic A, Gillman AN, Lauksund S, Robson Moe NA, Manzi A, Storflor M, Abel Zur Wiesch P, Abel S. RESTAMP - Rate estimates by sequence-tag analysis of microbial populations. Comput Struct Biotechnol J 2021; 19:1035-1051. [PMID: 33613869 PMCID: PMC7878984 DOI: 10.1016/j.csbj.2021.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/31/2022] Open
Abstract
Microbial division rates determine the speed of mutation accumulation and thus the emergence of antimicrobial resistance. Microbial death rates are affected by antibiotic action and the immune system. Therefore, measuring these rates has advanced our understanding of host-pathogen interactions and antibiotic action. Several methods based on marker-loss or few inheritable neutral markers exist that allow estimating microbial division and death rates, each of which has advantages and limitations. Technical bottlenecks, i.e., experimental sampling events, during the experiment can distort the rate estimates and are typically unaccounted for or require additional calibration experiments. In this work, we introduce RESTAMP (Rate Estimates by Sequence Tag Analysis of Microbial Populations) as a method for determining bacterial division and death rates. This method uses hundreds of fitness neutral sequence barcodes to measure the rates and account for experimental bottlenecks at the same time. We experimentally validate RESTAMP and compare it to established plasmid loss methods. We find that RESTAMP has a number of advantages over plasmid loss or previous marker based techniques. (i) It enables to correct the distortion of rate estimates by technical bottlenecks. (ii) Rate estimates are independent of the sequence tag distribution in the starting culture allowing the use of an arbitrary number of tags. (iii) It introduces a bottleneck sensitivity measure that can be used to maximize the accuracy of the experiment. RESTAMP allows studying microbial population dynamics with great resolution over a wide dynamic range and can thus advance our understanding of host-pathogen interactions or the mechanisms of antibiotic action.
Collapse
Affiliation(s)
- Anel Mahmutovic
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Aaron Nicholas Gillman
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA
| | - Silje Lauksund
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Natasha-Anne Robson Moe
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Aime Manzi
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Merete Storflor
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA
| | - Pia Abel Zur Wiesch
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, 0318 Oslo, Norway.,Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sören Abel
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, 0318 Oslo, Norway.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
14
|
Vlazaki M, Price DJ, Restif O. An experimental design tool to optimize inference precision in data-driven mathematical models of bacterial infections in vivo. J R Soc Interface 2020; 17:20200717. [PMID: 33323052 DOI: 10.1098/rsif.2020.0717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The management of bacterial diseases calls for a detailed knowledge about the dynamic changes in host-bacteria interactions. Biological insights are gained by integrating experimental data with mechanistic mathematical models to infer experimentally unobservable quantities. This inter-disciplinary field would benefit from experiments with maximal information content yielding high-precision inference. Here, we present a computationally efficient tool for optimizing experimental design in terms of parameter inference in studies using isogenic-tagged strains. We study the effect of three experimental design factors: number of biological replicates, sampling timepoint selection and number of copies per tagged strain. We conduct a simulation study to establish the relationship between our optimality criterion and the size of parameter estimate confidence intervals, and showcase its application in a range of biological scenarios reflecting different dynamics patterns observed in experimental infections. We show that in low-variance systems with low killing and replication rates, predicting high-precision experimental designs is consistently achieved; higher replicate sizes and strategic timepoint selection yield more precise estimates. Finally, we address the question of resource allocation under constraints; given a fixed number of host animals and a constraint on total inoculum size per host, infections with fewer strains at higher copies per strain lead to higher-precision inference.
Collapse
Affiliation(s)
- Myrto Vlazaki
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - David J Price
- Centre for Epidemiology and Biostatistics, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia.,The Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, Victoria 3000, Australia
| | - Olivier Restif
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| |
Collapse
|
15
|
De Ste Croix M, Holmes J, Wanford JJ, Moxon ER, Oggioni MR, Bayliss CD. Selective and non-selective bottlenecks as drivers of the evolution of hypermutable bacterial loci. Mol Microbiol 2020; 113:672-681. [PMID: 32185830 PMCID: PMC7154626 DOI: 10.1111/mmi.14453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/28/2022]
Abstract
Bottlenecks reduce the size of the gene pool within populations of all life forms with implications for their subsequent survival. Here, we examine the effects of bottlenecks on bacterial commensal-pathogens during transmission between, and dissemination within, hosts. By reducing genetic diversity, bottlenecks may alter individual or population-wide adaptive potential. A diverse range of hypermutable mechanisms have evolved in infectious agents that allow for rapid generation of genetic diversity in specific genomic loci as opposed to the variability arising from increased genome-wide mutation rates. These localised hypermutable mechanisms include multi-gene phase variation (PV) of outer membrane components, multi-allele PV of restriction systems and recombination-driven antigenic variation. We review selected experimental and theoretical (mathematical) models pertaining to the hypothesis that localised hypermutation (LH) compensates for fitness losses caused by bottlenecks and discuss whether bottlenecks have driven the evolution of hypermutable loci.
Collapse
Affiliation(s)
- Megan De Ste Croix
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Jonathan Holmes
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Joseph J Wanford
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - E Richard Moxon
- Department of Paediatrics, University of Oxford Medical Sciences Division, John Radcliffe Hospital, Oxford, UK
| | - Marco R Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | | |
Collapse
|
16
|
Vlazaki M, Huber J, Restif O. Integrating mathematical models with experimental data to investigate the within-host dynamics of bacterial infections. Pathog Dis 2020; 77:5704399. [PMID: 31942996 PMCID: PMC6986552 DOI: 10.1093/femspd/ftaa001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/13/2020] [Indexed: 12/23/2022] Open
Abstract
Bacterial infections still constitute a major cause of mortality and morbidity worldwide. The unavailability of therapeutics, antimicrobial resistance and the chronicity of infections due to incomplete clearance contribute to this phenomenon. Despite the progress in antimicrobial and vaccine development, knowledge about the effect that therapeutics have on the host–bacteria interactions remains incomplete. Insights into the characteristics of bacterial colonization and migration between tissues and the relationship between replication and host- or therapeutically induced killing can enable efficient design of treatment approaches. Recently, innovative experimental techniques have generated data enabling the qualitative characterization of aspects of bacterial dynamics. Here, we argue that mathematical modeling as an adjunct to experimental data can enrich the biological insight that these data provide. However, due to limited interdisciplinary training, efforts to combine the two remain limited. To promote this dialogue, we provide a categorization of modeling approaches highlighting their relationship to data generated by a range of experimental techniques in the area of in vivo bacterial dynamics. We outline common biological themes explored using mathematical models with case studies across all pathogen classes. Finally, this review advocates multidisciplinary integration to improve our mechanistic understanding of bacterial infections and guide the use of existing or new therapies.
Collapse
Affiliation(s)
- Myrto Vlazaki
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - John Huber
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - Olivier Restif
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| |
Collapse
|
17
|
Inhibition of invasive salmonella by orally administered IgA and IgG monoclonal antibodies. PLoS Negl Trop Dis 2020; 14:e0007803. [PMID: 32203503 PMCID: PMC7117778 DOI: 10.1371/journal.pntd.0007803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 04/02/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
Non-typhoidal Salmonella enterica strains, including serovar Typhimurium (STm), are an emerging cause of invasive disease among children and the immunocompromised, especially in regions of sub-Saharan Africa. STm invades the intestinal mucosa through Peyer's patch tissues before disseminating systemically. While vaccine development efforts are ongoing, the emergence of multidrug resistant strains of STm affirms the need to seek alternative strategies to protect high-risk individuals from infection. In this report, we investigated the potential of an orally administered O5 serotype-specific IgA monoclonal antibody (mAb), called Sal4, to prevent infection of invasive Salmonella enterica serovar Typhimurium (STm) in mice. Sal4 IgA was delivered to mice prior to or concurrently with STm challenge. Infectivity was measured as bacterial burden in Peyer's patch tissues one day after challenge. Using this model, we defined the minimal amount of Sal4 IgA required to significantly reduce STm uptake into Peyer's patches. The relative efficacy of Sal4 in dimeric and secretory IgA (SIgA) forms was compared. To assess the role of isotype in oral passive immunization, we engineered a recombinant IgG1 mAb carrying the Sal4 variable regions and evaluated its ability to block invasion of STm into epithelial cells in vitro and Peyer's patch tissues. Our results demonstrate the potential of orally administered monoclonal IgA and SIgA, but not IgG, to passively immunize against invasive Salmonella. Nonetheless, the prophylactic window of IgA/SIgA in the mouse was on the order of minutes, underscoring the need to develop formulations to protect mAbs in the gastric environment and to permit sustained release in the small intestine.
Collapse
|
18
|
Olivenza DR, Nicoloff H, Sánchez-Romero MA, Cota I, Andersson DI, Casadesús J. A portable epigenetic switch for bistable gene expression in bacteria. Sci Rep 2019; 9:11261. [PMID: 31375711 PMCID: PMC6677893 DOI: 10.1038/s41598-019-47650-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/22/2019] [Indexed: 02/04/2023] Open
Abstract
We describe a portable epigenetic switch based on opvAB, a Salmonella enterica operon that undergoes bistable expression under DNA methylation control. A DNA fragment containing the opvAB promoter and the opvAB upstream regulatory region confers bistability to heterologous genes, yielding OFF and ON subpopulations. Bistable expression under opvAB control is reproducible in Escherichia coli, showing that the opvAB switch can be functional in a heterologous host. Subpopulations of different sizes can be produced at will using engineered opvAB variants. Controlled formation of antibiotic-resistant and antibiotic-susceptible subpopulations may allow use of the opvAB switch in the study of bacterial heteroresistance to antibiotics.
Collapse
Affiliation(s)
- David R Olivenza
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Apartado 1095, 41080, Sevilla, Spain
| | - Hervé Nicoloff
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23, Uppsala, Sweden
| | | | - Ignacio Cota
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Apartado 1095, 41080, Sevilla, Spain.,Centre for Research in Agricultural Genomics, CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, 08193, Barcelona, Spain
| | - Dan I Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23, Uppsala, Sweden
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Apartado 1095, 41080, Sevilla, Spain.
| |
Collapse
|
19
|
Mutational and non mutational adaptation of Salmonella enterica to the gall bladder. Sci Rep 2019; 9:5203. [PMID: 30914708 PMCID: PMC6435676 DOI: 10.1038/s41598-019-41600-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
During systemic infection of susceptible hosts, Salmonella enterica colonizes the gall bladder, which contains lethal concentrations of bile salts. Recovery of Salmonella cells from the gall bladder of infected mice yields two types of isolates: (i) bile-resistant mutants; (ii) isolates that survive lethal selection without mutation. Bile-resistant mutants are recovered at frequencies high enough to suggest that increased mutation rates may occur in the gall bladder, thus providing a tentative example of stress-induced mutation in a natural environment. However, most bile-resistant mutants characterized in this study show defects in traits that are relevant for Salmonella colonization of the animal host. Mutation may thus permit short-term adaptation to the gall bladder at the expense of losing fitness for transmission to new hosts. In contrast, non mutational adaptation may have evolved as a fitness-preserving strategy. Failure of RpoS− mutants to colonize the gall bladder supports the involvement of the general stress response in non mutational adaptation.
Collapse
|
20
|
The Role of the Host in Driving Phenotypic Heterogeneity in Salmonella. Trends Microbiol 2019; 27:508-523. [PMID: 30755344 DOI: 10.1016/j.tim.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/21/2018] [Accepted: 01/18/2019] [Indexed: 02/07/2023]
Abstract
The complex infection environment within hosts exerts unique stresses across tissues and cell types, selecting for phenotypic heterogeneity in bacterial populations. Pathogens maintain variability during infection as a strategy to cope with fluctuating host immune conditions, leading to diversification of virulence phenotypes. Recent improvements in single-cell analyses have revealed that distinct bacterial subpopulations contribute unique colonization and growth strategies across infection sites. We discuss several examples of host-driven phenotypic heterogeneity in Salmonella populations throughout the course of infection, highlighting how variation in gene expression, growth rate, immune evasion, and metabolic activity contribute to overall bacterial success at the population level. We additionally focus our discussion on the implications of diversity within bacterial communities for antimicrobial efficacy.
Collapse
|
21
|
Proietti M, Perruzza L, Scribano D, Pellegrini G, D'Antuono R, Strati F, Raffaelli M, Gonzalez SF, Thelen M, Hardt WD, Slack E, Nicoletti M, Grassi F. ATP released by intestinal bacteria limits the generation of protective IgA against enteropathogens. Nat Commun 2019; 10:250. [PMID: 30651557 PMCID: PMC6335424 DOI: 10.1038/s41467-018-08156-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
T cell dependent secretory IgA (SIgA) generated in the Peyer's patches (PPs) of the small intestine shapes a broadly diverse microbiota that is crucial for host physiology. The mutualistic co-evolution of host and microbes led to the relative tolerance of host's immune system towards commensal microorganisms. The ATP-gated ionotropic P2X7 receptor limits T follicular helper (Tfh) cells expansion and germinal center (GC) reaction in the PPs. Here we show that transient depletion of intestinal ATP can dramatically improve high-affinity IgA response against both live and inactivated oral vaccines. Ectopic expression of Shigella flexneri periplasmic ATP-diphosphohydrolase (apyrase) abolishes ATP release by bacteria and improves the specific IgA response against live oral vaccines. Antibody responses primed in the absence of intestinal extracellular ATP (eATP) also provide superior protection from enteropathogenic infection. Thus, modulation of eATP in the small intestine can affect high-affinity IgA response against gut colonizing bacteria.
Collapse
Affiliation(s)
- Michele Proietti
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland.,CCI-Center for Chronic Immunodeficiency, Universitätsklinikum Freiburg, 79106, Freiburg, Germany
| | - Lisa Perruzza
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Daniela Scribano
- Department of Medical and Oral Sciences and Biotechnologies, University ''Gabriele D'Annunzio'', Via dei Vestini, Campus Universitario, 66100, Chieti, Italy.,Department of Public Health and Infectious Diseases, University ''La Sapienza'' of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057, Zurich, Switzerland
| | - Rocco D'Antuono
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Francesco Strati
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Marco Raffaelli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Emma Slack
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Mauro Nicoletti
- Department of Medical and Oral Sciences and Biotechnologies, University ''Gabriele D'Annunzio'', Via dei Vestini, Campus Universitario, 66100, Chieti, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Via Vincenzo Vela 6, 6500, Bellinzona, Switzerland. .,Istituto Nazionale Genetica Molecolare ''Romeo ed Enrica Invernizzi'', Via Francesco Sforza 35, 20122, Milan, Italy. .,Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Via Vanvitelli 32, 20129, Milan, Italy.
| |
Collapse
|
22
|
Sánchez-Romero MA, Casadesús J. Contribution of SPI-1 bistability to Salmonella enterica cooperative virulence: insights from single cell analysis. Sci Rep 2018; 8:14875. [PMID: 30291285 PMCID: PMC6173691 DOI: 10.1038/s41598-018-33137-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/20/2018] [Indexed: 11/25/2022] Open
Abstract
Salmonella enterica pathogenicity island 1 (SPI-1) is a gene cluster that encodes a type III secretion system and effectors involved in epithelial cell invasion. SPI-1 undergoes bistable expression, with concomitant formation of SPI-1ON and SPI-1OFF lineages. This study describes single cell analysis of SP1-1 bistability and epithelial cell invasion, and reports the unsuspected observation that optimal invasion of epithelial cells requires the presence of both SPI-1ON and SPI-1OFF subpopulations. The contribution of SPI-1OFF cells to optimal invasion may rely on their ability to invade epithelial cells if a SPI-1ON subpopulation is present. In fact, Salmonella SPI-1 mutants are also able to invade epithelial cells in the presence of SPI-1ONSalmonellae, a phenomenon described in the 1990’s by Galán and co-workers. Invasion by SPI-1OFF cells does not seem to involve a diffusible factor. A small number of SPI-1ON cells is sufficient to endow the bacterial population with invasion capacity, a feature that may permit host colonization regardless of the bottlenecks encountered by Salmonella populations inside animals.
Collapse
Affiliation(s)
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Apartado 1095, 41080, Sevilla, Spain
| |
Collapse
|
23
|
Kulkarni DH, McDonald KG, Knoop KA, Gustafsson JK, Kozlowski KM, Hunstad DA, Miller MJ, Newberry RD. Goblet cell associated antigen passages are inhibited during Salmonella typhimurium infection to prevent pathogen dissemination and limit responses to dietary antigens. Mucosal Immunol 2018; 11:1103-1113. [PMID: 29445136 PMCID: PMC6037413 DOI: 10.1038/s41385-018-0007-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/19/2017] [Accepted: 12/26/2017] [Indexed: 02/04/2023]
Abstract
Dietary antigen acquisition by lamina propria (LP) dendritic cells (DCs) is crucial to induce oral tolerance and maintain homeostasis. However, encountering innocuous antigens during infection can lead to inflammatory responses, suggesting processes may limit steady-state luminal antigen capture during infection. We observed that goblet cell (GC) associated antigen passages (GAPs), a steady-state pathway delivering luminal antigens to LP-DCs, are inhibited during Salmonella infection. GAP inhibition was mediated by IL-1β. Infection abrogated luminal antigen delivery and antigen-specific T cell proliferation in the mesenteric lymph node (MLN). Antigen-specific T cell proliferation to dietary antigen was restored by overriding GAP suppression; however, this did not restore regulatory T cell induction, but induced inflammatory T cell responses. Salmonella translocation to the MLN required GCs and correlated with GAPs. Genetic manipulations overriding GAP suppression, or antibiotics inducing colonic GAPs, but not antibiotics that do not, increased dissemination and worsened outcomes independent of luminal pathogen burden. Thus, steady-state sampling pathways are suppressed during infection to prevent responses to dietary antigens, limit pathogen entry, and lessen the disease. Moreover, antibiotics may worsen Salmonella infection by means beyond blunting gut microbiota colonization resistance, providing new insight into how precedent antibiotic use aggravates enteric infection.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Keely G McDonald
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Kathryn A Knoop
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jenny K Gustafsson
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Konrad M Kozlowski
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Mark J Miller
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Rodney D Newberry
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
24
|
Neutral barcoding of genomes reveals the dynamics of Salmonella colonization in cattle and their peripheral lymph nodes. Vet Microbiol 2018; 220:97-106. [PMID: 29885808 DOI: 10.1016/j.vetmic.2018.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022]
Abstract
Feedlot cattle often contain Salmonella. The number of bacteria that initiate colonization of different cattle organs and the bacterial migration within these large animals are poorly understood. To investigate these questions, we constructed wild-type isogenic tagged strains (WITS) of Salmonella by inserting 21-base barcodes flanked by Illumina sequencing primers into a neutral genome location. We then delivered several different pools of uniquely barcoded clones orally and into multiple intradermal sites, in individual Holstein steers, and subsequently performed Salmonella-directed sequence tag-based analysis of microbial populations (STAMP). Using high-throughput sequencing of the barcodes of Salmonella grown from steer lymph nodes, organs and feces, we monitored how individual barcoded clones travel from different entry sites within animals. Data showed that gastrointestinal colonization was established by up to hundreds of Salmonella founder cells, whereas peripheral lymph nodes were usually colonized by very low numbers of founding bacteria, often originating from the nearest draining intradermal delivery site. Transmission of Salmonella from the gastrointestinal tract to the lymphatic system was frequently observed, whereas entry of intradermally delivered bacteria into the gut was rare. Bacteria undergo limited extraintestinal proliferation within or prior to arrival at peripheral lymph nodes. Overall, the application of the STAMP technique facilitated characterization of the migration routes and founder population size of Salmonella within feedlot cattle and their organs and lymph nodes in unprecedented detail.
Collapse
|
25
|
Solanki V, Tiwari M, Tiwari V. Host-bacteria interaction and adhesin study for development of therapeutics. Int J Biol Macromol 2018; 112:54-64. [PMID: 29414732 DOI: 10.1016/j.ijbiomac.2018.01.151] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/15/2022]
Abstract
Host-pathogen interaction is one of the most important areas of study to understand the adhesion of the pathogen to the host organisms. To adhere on the host cell surface, bacteria assemble the diverse adhesive structures on its surface, which play a foremost role in targeting to the host cell. We have highlighted different bacterial adhesins which are either protein mediated or glycan mediated. The present article listed examples of different bacterial adhesin proteins involved in the interactions with their host, types and subtypes of the fimbriae and non-fimbriae bacterial adhesins. Different bacterial surface adhesin subunits interact with host via different host surface biomolecules. We have also discussed the interactome of some of the pathogens with their host. Therefore, the present study will help researchers to have a detailed understanding of different interacting bacterial adhesins and henceforth, develop new therapies, adhesin specific antibodies and vaccines, which can effectively control pathogenicity of the pathogens.
Collapse
Affiliation(s)
- Vandana Solanki
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India.
| |
Collapse
|
26
|
Price DJ, Breuzé A, Dybowski R, Mastroeni P, Restif O. An efficient moments-based inference method for within-host bacterial infection dynamics. PLoS Comput Biol 2017; 13:e1005841. [PMID: 29155811 PMCID: PMC5714343 DOI: 10.1371/journal.pcbi.1005841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 12/04/2017] [Accepted: 10/22/2017] [Indexed: 11/18/2022] Open
Abstract
Over the last ten years, isogenic tagging (IT) has revolutionised the study of bacterial infection dynamics in laboratory animal models. However, quantitative analysis of IT data has been hindered by the piecemeal development of relevant statistical models. The most promising approach relies on stochastic Markovian models of bacterial population dynamics within and among organs. Here we present an efficient numerical method to fit such stochastic dynamic models to in vivo experimental IT data. A common approach to statistical inference with stochastic dynamic models relies on producing large numbers of simulations, but this remains a slow and inefficient method for all but simple problems, especially when tracking bacteria in multiple locations simultaneously. Instead, we derive and solve the systems of ordinary differential equations for the two lower-order moments of the stochastic variables (mean, variance and covariance). For any given model structure, and assuming linear dynamic rates, we demonstrate how the model parameters can be efficiently and accurately estimated by divergence minimisation. We then apply our method to an experimental dataset and compare the estimates and goodness-of-fit to those obtained by maximum likelihood estimation. While both sets of parameter estimates had overlapping confidence regions, the new method produced lower values for the division and death rates of bacteria: these improved the goodness-of-fit at the second time point at the expense of that of the first time point. This flexible framework can easily be applied to a range of experimental systems. Its computational efficiency paves the way for model comparison and optimal experimental design.
Collapse
Affiliation(s)
- David J. Price
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Alexandre Breuzé
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- ENSTA-ParisTech, Palaiseau, France
| | - Richard Dybowski
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Piero Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Olivier Restif
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Inferring within-host bottleneck size: A Bayesian approach. J Theor Biol 2017; 435:218-228. [PMID: 28919397 DOI: 10.1016/j.jtbi.2017.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 08/07/2017] [Accepted: 09/13/2017] [Indexed: 12/21/2022]
Abstract
Recent technical developments in microbiology have led to new discoveries on the within-host dynamics of bacterial infections in laboratory animals. In particular, they have highlighted the importance of stochastic bottlenecks at the onset of invasive disease. A number of approaches exist for bottleneck-size estimation with respect to within-host bacterial infections; however, some are more appropriate than others under certain circumstances. A Bayesian comparison of several approaches is made in terms of the availability of isogenic multitype bacteria (e.g., WITS), knowledge of post-bottleneck dynamics, and the suitability of dilution with monotype bacteria. A sampling approach to bottleneck-size estimation is also introduced. The results are summarised by a guiding flowchart, which we hope will promote the use of quantitative models in microbiology to refine the analysis of animal experiment data.
Collapse
|
28
|
Kim D, Seo SU, Zeng MY, Kim WU, Kamada N, Inohara N, Núñez G. Mesenchymal Cell-Specific MyD88 Signaling Promotes Systemic Dissemination of Salmonella Typhimurium via Inflammatory Monocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1362-1371. [PMID: 28674182 PMCID: PMC5548622 DOI: 10.4049/jimmunol.1601527] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 06/07/2017] [Indexed: 12/24/2022]
Abstract
Enteric pathogens including Salmonella enteric serovar Typhimurium can breach the epithelial barrier of the host and spread to systemic tissues. In response to infection, the host activates innate immune receptors via the signaling molecule MyD88, which induces protective inflammatory and antimicrobial responses. Most of these innate immune responses have been studied in hematopoietic cells, but the role of MyD88 signaling in other cell types remains poorly understood. Surprisingly, we found that Dermo1-Cre;Myd88fl/fl mice with mesenchymal cell-specific deficiency of MyD88 were less susceptible to orogastric and i.p. STyphimurium infection than their Myd88fl/fl littermates. The reduced susceptibility of Dermo1-Cre;Myd88fl/fl mice to infection was associated with lower loads of S. Typhimurium in the liver and spleen. Mutant analyses revealed that S. Typhimurium employs its virulence type III secretion system 2 to promote its growth through MyD88 signaling pathways in mesenchymal cells. Inflammatory monocytes function as a major cell population for systemic dissemination of S. Typhimurium Mechanistically, mesenchymal cell-specific MyD88 signaling promoted CCL2 production in the liver and spleen and recruitment of inflammatory monocytes to systemic organs in response to STyphimurium infection. Consistently, MyD88 signaling in mesenchymal cells enhanced the number of phagocytes including Ly6ChiLy6G- inflammatory monocytes harboring STyphimurium in the liver. These results suggest that S. Typhimurium promotes its systemic growth and dissemination through MyD88 signaling pathways in mesenchymal cells.
Collapse
Affiliation(s)
- Donghyun Kim
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, Catholic University of Korea, Seoul 06591, Korea
| | - Sang-Uk Seo
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
- Department of Biomedical Sciences, Wide River Institute of Immunology, Seoul National University College of Medicine, Gangwon-do 25159, Korea
| | - Melody Y Zeng
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, Catholic University of Korea, Seoul 06591, Korea
- Department of Internal Medicine, College of Medicine, Catholic University of Korea, Seoul 06591, Korea; and
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Naohiro Inohara
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Gabriel Núñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109;
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
29
|
Bumann D, Schothorst J. Intracellular Salmonella metabolism. Cell Microbiol 2017; 19. [PMID: 28672057 DOI: 10.1111/cmi.12766] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 12/28/2022]
Abstract
Growth of Salmonella inside infected host cells is a key aspect of their ability to cause local enteritis or systemic disease. This growth depends on exploitation of host nutrients through a large Salmonella metabolism network with hundreds of metabolites and enzymes. Studies in cell culture infection models are unravelling more and more of the underlying molecular and cellular mechanisms but also show striking Salmonella metabolic plasticity depending on host cell line and experimental conditions. In vivo studies have revealed a qualitatively diverse, but quantitatively poor, host-Salmonella nutritional interface, which on one side makes Salmonella fitness largely resilient against metabolic perturbations, but on the other side severely limits Salmonella biomass generation and growth rates. This review discusses goals and techniques for studying Salmonella intracellular metabolism, summarises main results and implications, and proposes key issues that could be addressed in future studies.
Collapse
Affiliation(s)
- Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Joep Schothorst
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
30
|
Deciphering the landscape of host barriers to Listeria monocytogenes infection. Proc Natl Acad Sci U S A 2017; 114:6334-6339. [PMID: 28559314 DOI: 10.1073/pnas.1702077114] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Listeria monocytogenes is a common food-borne pathogen that can disseminate from the intestine and infect multiple organs. Here, we used sequence tag-based analysis of microbial populations (STAMP) to investigate Lmonocytogenes population dynamics during infection. We created a genetically barcoded library of murinized Lmonocytogenes and then used deep sequencing to track the pathogen's dissemination routes and quantify its founding population (Nb) sizes in different organs. We found that the pathogen disseminates from the gastrointestinal tract to distal sites through multiple independent routes and that Nb sizes vary greatly among tissues, indicative of diverse host barriers to infection. Unexpectedly, comparative analyses of sequence tags revealed that fecally excreted organisms are largely derived from the very small number of L. monocytogenes cells that colonize the gallbladder. Immune depletion studies suggest that distinct innate immune cells restrict the pathogen's capacity to establish replicative niches in the spleen and liver. Finally, studies in germ-free mice suggest that the microbiota plays a critical role in the development of the splenic, but not the hepatic, barriers that prevent L. monocytogenes from seeding these organs. Collectively, these observations illustrate the potency of the STAMP approach to decipher the impact of host factors on population dynamics of pathogens during infection.
Collapse
|
31
|
Berthelot JM, Claudepierre P. Trafficking of antigens from gut to sacroiliac joints and spine in reactive arthritis and spondyloarthropathies: Mainly through lymphatics? Joint Bone Spine 2016; 83:485-90. [DOI: 10.1016/j.jbspin.2015.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022]
|
32
|
A Bioluminescent Francisella tularensis SCHU S4 Strain Enables Noninvasive Tracking of Bacterial Dissemination and the Evaluation of Antibiotics in an Inhalational Mouse Model of Tularemia. Antimicrob Agents Chemother 2016; 60:7206-7215. [PMID: 27671061 DOI: 10.1128/aac.01586-16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/08/2016] [Indexed: 11/20/2022] Open
Abstract
Bioluminescence imaging (BLI) enables real-time, noninvasive tracking of infection in vivo and longitudinal infection studies. In this study, a bioluminescent Francisella tularensis strain, SCHU S4-lux, was used to develop an inhalational infection model in BALB/c mice. Mice were infected intranasally, and the progression of infection was monitored in real time using BLI. A bioluminescent signal was detectable from 3 days postinfection (3 dpi), initially in the spleen and then in the liver and lymph nodes, before finally becoming systemic. The level of bioluminescent signal correlated with bacterial numbers in vivo, enabling noninvasive quantification of bacterial burdens in tissues. Treatment with levofloxacin (commencing at 4 dpi) significantly reduced the BLI signal. Furthermore, BLI was able to distinguish noninvasively between different levofloxacin treatment regimens and to identify sites of relapse following treatment cessation. These data demonstrate that BLI and SCHU S4-lux are suitable for the study of F. tularensis pathogenesis and the evaluation of therapeutics for tularemia.
Collapse
|
33
|
Abstract
Recent developments in studies of tumor heterogeneity have provoked new thoughts on cancer management. There is a desperate need to understand influence of the tumor microenvironment on cancer development and evolution. Applying principles and quantitative methods from ecology can suggest novel solutions to fulfil this need. We discuss spatial heterogeneity as a fundamental biological feature of the microenvironment, which has been largely ignored. Histological samples can provide spatial context of diverse cell types coexisting within the microenvironment. Advanced computer-vision techniques have been developed for spatial mapping of cells in histological samples. This has enabled the applications of experimental and analytical tools from ecology to cancer research, generating system-level knowledge of microenvironmental spatial heterogeneity. We focus on studies of immune infiltrate and tumor resource distribution, and highlight statistical approaches for addressing the emerging challenges based on these new approaches.
Collapse
Affiliation(s)
- Yinyin Yuan
- Centre for Evolution and Cancer and Division of Molecular Pathology, The Institute of Cancer Research, London; and Centre for Molecular Pathology, Royal Marsden Hospital, London
| |
Collapse
|
34
|
Spadoni I, Zagato E, Bertocchi A, Paolinelli R, Hot E, Di Sabatino A, Caprioli F, Bottiglieri L, Oldani A, Viale G, Penna G, Dejana E, Rescigno M. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015; 350:830-4. [PMID: 26564856 DOI: 10.1126/science.aad0135] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In healthy individuals, the intestinal microbiota cannot access the liver, spleen, or other peripheral tissues. Some pathogenic bacteria can reach these sites, however, and can induce a systemic immune response. How such compartmentalization is achieved is unknown. We identify a gut-vascular barrier (GVB) in mice and humans that controls the translocation of antigens into the blood stream and prohibits entry of the microbiota. Salmonella typhimurium can penetrate the GVB in a manner dependent on its pathogenicity island (Spi) 2-encoded type III secretion system and on decreased β-catenin-dependent signaling in gut endothelial cells. The GVB is modified in celiac disease patients with elevated serum transaminases, which indicates that GVB dismantling may be responsible for liver damage in these patients. Understanding the GVB may provide new insights into the regulation of the gut-liver axis.
Collapse
Affiliation(s)
- Ilaria Spadoni
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Elena Zagato
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alice Bertocchi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Roberta Paolinelli
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Edina Hot
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Antonio Di Sabatino
- First Department of Medicine, St. Matteo Hospital, University of Pavia, Pavia, Italy
| | - Flavio Caprioli
- Unità Operativa Gastroenterologia ed Endoscopia, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico di Milano, and Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milan, Italy
| | - Luca Bottiglieri
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Amanda Oldani
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Giuseppe Viale
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Giuseppe Penna
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Elisabetta Dejana
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy. Department of Biosciences, Università degli Studi di Milano, Italy. Department of Genetics, Immunology and Pathology, Uppsala University, Uppsala, Sweden
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy. Department of Biosciences, Università degli Studi di Milano, Italy.
| |
Collapse
|
35
|
Identification of Population Bottlenecks and Colonization Factors during Assembly of Bacterial Communities within the Zebrafish Intestine. mBio 2015; 6:e01163-15. [PMID: 26507229 PMCID: PMC4626852 DOI: 10.1128/mbio.01163-15] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The zebrafish, Danio rerio, is a powerful model for studying bacterial colonization of the vertebrate intestine, but the genes required by commensal bacteria to colonize the zebrafish gut have not yet been interrogated on a genome-wide level. Here we apply a high-throughput transposon mutagenesis screen to Aeromonas veronii Hm21 and Vibrio sp. strain ZWU0020 during their colonization of the zebrafish intestine alone and in competition with each other, as well as in different colonization orders. We use these transposon-tagged libraries to track bacterial population sizes in different colonization regimes and to identify gene functions required during these processes. We show that intraspecific, but not interspecific, competition with a previously established bacterial population greatly reduces the ability of these two bacterial species to colonize. Further, using a simple binomial sampling model, we show that under conditions of interspecific competition, genes required for colonization cannot be identified because of the population bottleneck experienced by the second colonizer. When bacteria colonize the intestine alone or at the same time as the other species, we find shared suites of functional requirements for colonization by the two species, including a prominent role for chemotaxis and motility, regardless of the presence of another species. Zebrafish larvae, which are amenable to large-scale gnotobiotic studies, comprehensive sampling of their intestinal microbiota, and live imaging, are an excellent model for investigations of vertebrate intestinal colonization dynamics. We sought to develop a mutagenesis and tagging system in order to understand bacterial population dynamics and functional requirements during colonization of the larval zebrafish intestine. We explored changes in bacterial colonization dynamics and functional requirements when bacteria colonize a bacterium-free intestine, one previously colonized by their own species, or one colonized previously or simultaneously with a different species. This work provides a framework for rapid identification of colonization factors important under different colonization conditions. Furthermore, we demonstrate that when colonizing bacterial populations are very small, this approach is not accurate because random sampling of the input pool is sufficient to explain the distribution of inserts recovered from bacteria that colonized the intestines.
Collapse
|
36
|
O'Donnell H, Pham OH, Benoun JM, Ravesloot-Chávez MM, McSorley SJ. Contaminated water delivery as a simple and effective method of experimental Salmonella infection. Future Microbiol 2015; 10:1615-27. [PMID: 26439708 DOI: 10.2217/fmb.15.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIM In most infectious disease models, it is assumed that gavage needle infection is the most reliable means of pathogen delivery to the GI tract. However, this methodology can cause esophageal tearing and induces stress in experimental animals, both of which have the potential to impact early infection and the subsequent immune response. MATERIALS & METHODS C57BL/6 mice were orally infected with virulent Salmonella Typhimurium SL1344 either by intragastric gavage preceded by sodium bicarbonate, or by contamination of drinking water. RESULTS We demonstrate that water contamination delivery of Salmonella is equivalent to gavage inoculation in providing a consistent model of infection. Furthermore, exposure of mice to contaminated drinking water for as little as 4 h allowed maximal mucosal and systemic infection, suggesting an abbreviated window exists for natural intestinal entry. CONCLUSION Together, these data question the need for gavage delivery for infection with oral pathogens.
Collapse
Affiliation(s)
- Hope O'Donnell
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA.,Yersinia Research Unit, Microbiology Department, Pasteur Institute, Paris, France
| | - Oanh H Pham
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Joseph M Benoun
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Marietta M Ravesloot-Chávez
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
37
|
Ziętara N, Łyszkiewicz M, Puchałka J, Witzlau K, Reinhardt A, Förster R, Pabst O, Prinz I, Krueger A. Multicongenic fate mapping quantification of dynamics of thymus colonization. ACTA ACUST UNITED AC 2015; 212:1589-601. [PMID: 26347471 PMCID: PMC4577840 DOI: 10.1084/jem.20142143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/10/2015] [Indexed: 12/30/2022]
Abstract
Ziętara et al demonstrate with multicongenic fate mapping that thymus seeding is directly restricted to the duration of niche occupancy rather than long-range effects. Postnatal T cell development depends on continuous colonization of the thymus by BM-derived T lineage progenitors. Both quantitative parameters and the mechanisms of thymus seeding remain poorly understood. Here, we determined the number of dedicated thymus-seeding progenitor niches (TSPNs) capable of supporting productive T cell development, turnover rates of niche occupancy, and feedback mechanisms. To this end, we established multicongenic fate mapping combined with mathematical modeling to quantitate individual events of thymus colonization. We applied this method to study thymus colonization in CCR7−/−CCR9−/− (DKO) mice, whose TSPNs are largely unoccupied. We showed that ∼160–200 TSPNs are present in the adult thymus and, on average, 10 of these TSPNs were open for recolonization at steady state. Preconditioning of wild-type mice revealed a similar number of TSPNs, indicating that preconditioning can generate space efficiently for transplanted T cell progenitors. To identify potential cellular feedback loops restricting thymus colonization, we performed serial transfer experiments. These experiments indicated that thymus seeding was directly restricted by the duration of niche occupancy rather than long-range effects, thus challenging current paradigms of thymus colonization.
Collapse
Affiliation(s)
- Natalia Ziętara
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Marcin Łyszkiewicz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Jacek Puchałka
- Dr. von Haunersches Kinderspital, University Children's Hospital, Ludwig Maximilian University, D-80337 Munich, Germany
| | - Katrin Witzlau
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Annika Reinhardt
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Oliver Pabst
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany Institute of Molecular Medicine, RWTH Aachen University, D-52074 Aachen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Andreas Krueger
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
38
|
Kreibich S, Hardt WD. Experimental approaches to phenotypic diversity in infection. Curr Opin Microbiol 2015; 27:25-36. [PMID: 26143306 DOI: 10.1016/j.mib.2015.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 06/03/2015] [Accepted: 06/06/2015] [Indexed: 12/16/2022]
Abstract
Microbial infections are burdening human health, even after the advent of antibiotics, vaccines and hygiene. Thus, infection biology has aimed at the molecular understanding of the pathogen-host interaction. This has revealed key virulence factors, host cell signaling pathways and immune responses. However, our understanding of the infection process is still incomplete. Recent evidence suggests that phenotypic diversity can have important consequences for the infection process. Diversity arises from the formation of distinct subpopulations of pathogen cells (with distinct virulence factor expression patterns) and host cells (with distinct response capacities). For technical reasons, such phenotypic diversity has often been overlooked. We are highlighting several striking examples and discuss the experimental approaches available for analyzing the different subpopulations. Single cell reporters and approaches from systems biology do hold much promise.
Collapse
Affiliation(s)
- Saskia Kreibich
- Institute of Microbiology, ETH Zürich, CH-8093 Zürich, Switzerland
| | | |
Collapse
|
39
|
Abel S, Abel zur Wiesch P, Davis BM, Waldor MK. Analysis of Bottlenecks in Experimental Models of Infection. PLoS Pathog 2015; 11:e1004823. [PMID: 26066486 PMCID: PMC4465827 DOI: 10.1371/journal.ppat.1004823] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Sören Abel
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
- Department of Pharmacy, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
- * E-mail: (SA); (MKW)
| | - Pia Abel zur Wiesch
- Department of Pharmacy, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
- Division of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Brigid M. Davis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
| | - Matthew K. Waldor
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
- * E-mail: (SA); (MKW)
| |
Collapse
|
40
|
Intracellular Listeria monocytogenes comprises a minimal but vital fraction of the intestinal burden following foodborne infection. Infect Immun 2015; 83:3146-56. [PMID: 26015479 DOI: 10.1128/iai.00503-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022] Open
Abstract
Listeria monocytogenes is a highly adaptive bacterium that replicates as a free-living saprophyte in the environment as well as a facultative intracellular pathogen that causes invasive foodborne infections. The intracellular life cycle of L. monocytogenes is considered to be its primary virulence determinant during mammalian infection; however, the proportion of L. monocytogenes that is intracellular in vivo has not been studied extensively. In this report, we demonstrate that the majority of wild-type (strain EGDe) and mouse-adapted (InlA(m)-expressing) L. monocytogenes recovered from the mesenteric lymph nodes (MLN) was extracellular within the first few days after foodborne infection. In addition, significantly lower burdens of L. monocytogenes were recovered from the colon, spleen, and liver of gentamicin-treated mice than of control mice. This led us to investigate whether intracellular replication of L. monocytogenes was essential during the intestinal phase of infection. We found that lipoate protein ligase-deficient L. monocytogenes (ΔlplA1) mutants, which display impaired intracellular growth, were able to colonize the colon but did not persist efficiently and had a significant defect in spreading to the MLN, spleen, and liver. Together, these data indicate that the majority of the L. monocytogenes burden in the gastrointestinal tract is extracellular, but the small proportion of intracellular L. monocytogenes is essential for dissemination to the MLN and systemic organs.
Collapse
|
41
|
Sequence tag-based analysis of microbial population dynamics. Nat Methods 2015; 12:223-6, 3 p following 226. [PMID: 25599549 PMCID: PMC4344388 DOI: 10.1038/nmeth.3253] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/10/2014] [Indexed: 01/20/2023]
Abstract
We describe sequence tag-based analysis of microbial populations (STAMP) for characterization of pathogen population dynamics during infection. STAMP analyzes the frequency changes of genetically 'barcoded' organisms to quantify population bottlenecks and infer the founding population size. Analyses of intraintestinal Vibrio cholerae revealed infection-stage and region-specific host barriers to infection and showed unexpected V. cholerae migration counter to intestinal flow. STAMP provides a robust, widely applicable analytical framework for high-confidence characterization of in vivo microbial dissemination.
Collapse
|
42
|
Maier L, Diard M, Sellin ME, Chouffane ES, Trautwein-Weidner K, Periaswamy B, Slack E, Dolowschiak T, Stecher B, Loverdo C, Regoes RR, Hardt WD. Granulocytes impose a tight bottleneck upon the gut luminal pathogen population during Salmonella typhimurium colitis. PLoS Pathog 2014; 10:e1004557. [PMID: 25522364 PMCID: PMC4270771 DOI: 10.1371/journal.ppat.1004557] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/06/2014] [Indexed: 12/22/2022] Open
Abstract
Topological, chemical and immunological barriers are thought to limit infection by enteropathogenic bacteria. However, in many cases these barriers and their consequences for the infection process remain incompletely understood. Here, we employed a mouse model for Salmonella colitis and a mixed inoculum approach to identify barriers limiting the gut luminal pathogen population. Mice were infected via the oral route with wild type S. Typhimurium (S. Tm) and/or mixtures of phenotypically identical but differentially tagged S. Tm strains ("WITS", wild-type isogenic tagged strains), which can be individually tracked by quantitative real-time PCR. WITS dilution experiments identified a substantial loss in tag/genetic diversity within the gut luminal S. Tm population by days 2-4 post infection. The diversity-loss was not attributable to overgrowth by S. Tm mutants, but required inflammation, Gr-1+ cells (mainly neutrophilic granulocytes) and most likely NADPH-oxidase-mediated defense, but not iNOS. Mathematical modelling indicated that inflammation inflicts a bottleneck transiently restricting the gut luminal S. Tm population to approximately 6000 cells and plating experiments verified a transient, inflammation- and Gr-1+ cell-dependent dip in the gut luminal S. Tm population at day 2 post infection. We conclude that granulocytes, an important clinical hallmark of S. Tm-induced inflammation, impose a drastic bottleneck upon the pathogen population. This extends the current view of inflammation-fuelled gut-luminal Salmonella growth by establishing the host response in the intestinal lumen as a double-edged sword, fostering and diminishing colonization in a dynamic equilibrium. Our work identifies a potent immune defense against gut infection and reveals a potential Achilles' heel of the infection process which might be targeted for therapy.
Collapse
Affiliation(s)
- Lisa Maier
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Médéric Diard
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Mikael E. Sellin
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Elsa-Sarah Chouffane
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | | | - Balamurugan Periaswamy
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Emma Slack
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Tamas Dolowschiak
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| | - Bärbel Stecher
- Max von Pettenkofer-Institut, München, Germany; German Center for Infection Research (DZIF), partner site Ludwig Maximilian University of Munich, Munich, Germany
| | - Claude Loverdo
- Eidgenössische Technische Hochschule Zürich, Institute of Integrative Biology, Zurich, Switzerland
| | - Roland R. Regoes
- Eidgenössische Technische Hochschule Zürich, Institute of Integrative Biology, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Eidgenössische Technische Hochschule Zürich, Institute of Microbiology, Zurich, Switzerland
| |
Collapse
|
43
|
Lam LH, Monack DM. Intraspecies competition for niches in the distal gut dictate transmission during persistent Salmonella infection. PLoS Pathog 2014; 10:e1004527. [PMID: 25474319 PMCID: PMC4256465 DOI: 10.1371/journal.ppat.1004527] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 10/16/2014] [Indexed: 12/31/2022] Open
Abstract
In order to be transmitted, a pathogen must first successfully colonize and multiply within a host. Ecological principles can be applied to study host-pathogen interactions to predict transmission dynamics. Little is known about the population biology of Salmonella during persistent infection. To define Salmonella enterica serovar Typhimurium population structure in this context, 129SvJ mice were oral gavaged with a mixture of eight wild-type isogenic tagged Salmonella (WITS) strains. Distinct subpopulations arose within intestinal and systemic tissues after 35 days, and clonal expansion of the cecal and colonic subpopulation was responsible for increases in Salmonella fecal shedding. A co-infection system utilizing differentially marked isogenic strains was developed in which each mouse received one strain orally and the other systemically by intraperitoneal (IP) injection. Co-infections demonstrated that the intestinal subpopulation exerted intraspecies priority effects by excluding systemic S. Typhimurium from colonizing an extracellular niche within the cecum and colon. Importantly, the systemic strain was excluded from these distal gut sites and was not transmitted to naïve hosts. In addition, S. Typhimurium required hydrogenase, an enzyme that mediates acquisition of hydrogen from the gut microbiota, during the first week of infection to exert priority effects in the gut. Thus, early inhibitory priority effects are facilitated by the acquisition of nutrients, which allow S. Typhimurium to successfully compete for a nutritional niche in the distal gut. We also show that intraspecies colonization resistance is maintained by Salmonella Pathogenicity Islands SPI1 and SPI2 during persistent distal gut infection. Thus, important virulence effectors not only modulate interactions with host cells, but are crucial for Salmonella colonization of an extracellular intestinal niche and thereby also shape intraspecies dynamics. We conclude that priority effects and intraspecies competition for colonization niches in the distal gut control Salmonella population assembly and transmission.
Collapse
Affiliation(s)
- Lilian H. Lam
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Denise M. Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|