1
|
Xu J, Li P, Li Z, Liu S, Guo H, Lesser CF, Ke J, Zhao W, Mou X. Gut bacterial type III secretion systems aggravate colitis in mice and serve as biomarkers of Crohn's disease. EBioMedicine 2024; 107:105296. [PMID: 39216231 PMCID: PMC11402190 DOI: 10.1016/j.ebiom.2024.105296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Mesenteric adipose tissue (mAT) hyperplasia, known as creeping fat, is a pathologic characteristic of Crohn's disease (CD). In our previously reported cohort, we observed that Achromobacter pulmonis was the most abundant and prevalent bacteria cultivated from creeping fat. METHODS A whole genomic sequencing and identification of T3SS orthologs of mAT-derived A. pulmonis were used. A functional type III secretion system (T3SS) mediated the pathogenic potential of A. pulmonis in vitro and in mouse colitis model. Furthermore, a T3SS Finder pipeline was introduced to evaluate gut bacterial T3SS orthologs in the feces of CD patients, ulcerative colitis and colorectal cancer patients. FINDINGS Here, we reveal that mAT-derived A. pulmonis possesses a functional T3SS, aggravates colitis in mice via T3SS, and exhibits T3SS-dependent cytotoxicity via a caspase-independent mechanism in macrophages and epithelial cells, which demonstrated the pathogenic potential of the T3SS-harboring A. pulmonis. Metagenomic analyses demonstrate an increased abundance of Achromobacter in the fecal of Crohn's disease patients compared to healthy controls. A comprehensive comparison of total microbial vT3SS abundance in various intestine diseases demonstrated that the specific enrichment of vT3SS genes was shown in fecal samples of CD, neither ulcerative colitis nor colorectal cancer patients, and ten T3SS gene-based biomarkers for CD were discovered and validated in a newly recruited CD cohort. Furthermore, treatment with exclusive enteral nutrition (EEN), an intervention that improves CD patient symptomatology, was found associated with a significant reduction in the prevalence of T3SS genes in fecal samples. INTERPRETATION These findings highlight the pathogenic significance of T3SSs in the context of CD and identify specific T3SS genes that could potentially function as biomarkers for diagnosing and monitoring the clinical status of CD patients. FUNDING This work is supported by the National Key Research and Development Program of China (2020YFA0907800), the China Postdoctoral Science Foundation (2023M744089), the National Natural Science Foundation of China (32000096), the Shenzhen Science and Technology Programs (KQTD20200820145822023, RCIC20231211085944057, and ZDSYS20220606100803007), National Key Clinical Discipline, Guangdong Provincial Clinical Research Center for Digestive Diseases (2020B1111170004), Qingfeng Scientific Research Fund of the China Crohn's & Colitis Foundation (CCCF) (CCCF-QF-2022B71-1), and the Sixth Affiliated Hospital, Sun Yat-sen University Clinical Research 1010 Program 1010CG(2023)-08. These funding provided well support for this research work, which involved data collection, analysis, interpretation, patient recruitment and so on.
Collapse
Affiliation(s)
- Jun Xu
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Peijie Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhenye Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Sheng Liu
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Huating Guo
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jia Ke
- Department of General Surgery (Intestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Wenjing Zhao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Xiangyu Mou
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| |
Collapse
|
2
|
Yang R, Atkinson S, Chen Z, Cui Y, Du Z, Han Y, Sebbane F, Slavin P, Song Y, Yan Y, Wu Y, Xu L, Zhang C, Zhang Y, Hinnebusch BJ, Stenseth NC, Motin VL. Yersinia pestis and Plague: some knowns and unknowns. ZOONOSES (BURLINGTON, MASS.) 2023; 3:5. [PMID: 37602146 PMCID: PMC10438918 DOI: 10.15212/zoonoses-2022-0040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Since its first identification in 1894 during the third pandemic in Hong Kong, there has been significant progress of understanding the lifestyle of Yersinia pestis, the pathogen that is responsible for plague. Although we now have some understanding of the pathogen's physiology, genetics, genomics, evolution, gene regulation, pathogenesis and immunity, there are many unknown aspects of the pathogen and its disease development. Here, we focus on some of the knowns and unknowns relating to Y. pestis and plague. We notably focus on some key Y. pestis physiological and virulence traits that are important for its mammal-flea-mammal life cycle but also its emergence from the enteropathogen Yersinia pseudotuberculosis. Some aspects of the genetic diversity of Y. pestis, the distribution and ecology of plague as well as the medical countermeasures to protect our population are also provided. Lastly, we present some biosafety and biosecurity information related to Y. pestis and plague.
Collapse
Affiliation(s)
- Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Steve Atkinson
- School of Life Sciences, Centre for Biomolecular Science, University of Nottingham, Nottingham, United Kingdom
| | - Ziqi Chen
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Yujun Cui
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Zongmin Du
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yanping Han
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Florent Sebbane
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Philip Slavin
- Division of History and Politics, University of Stirling, Stirling FK9 4LJ, UK
| | - Yajun Song
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yanfeng Yan
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yarong Wu
- Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Lei Xu
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - Chutian Zhang
- College of Natural Resources and Environment, Northwest A&F University, Yangling 712100, China
| | - Yun Zhang
- Vanke School of Public Health, Tsinghua University, Beijing 100084, China
| | - B. Joseph Hinnebusch
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | - Nils Chr. Stenseth
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, N-0316 Oslo, Norway
| | - Vladimir L. Motin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
3
|
Single-cell transcriptomics of immune cells in lymph nodes reveals their composition and alterations in functional dynamics during the early stages of bubonic plague. SCIENCE CHINA. LIFE SCIENCES 2023; 66:110-126. [PMID: 35943690 DOI: 10.1007/s11427-021-2119-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Bubonic plague caused by Yersinia pestis is highly infectious and often fatal. Characterization of the host immune response and its subsequent suppression by Y. pestis is critical to understanding the pathogenesis of Y. pestis. Here, we utilized single-cell RNA sequencing to systematically profile the transcriptomes of immune cells in draining lymph nodes (dLNs) during the early stage of Y. pestis infection. Dendritic cells responded to Y. pestis within 2 h post-infection (hpi), followed by the activation of macrophages/monocytes (Mφs/Mons) and recruitment of polymorphonuclear neutrophils (PMNs) to dLNs at 24 hpi. Analysis of cell-to-cell communication suggests that PMNs may be recruited to lymph nodes following the secretion of CCL9 by Mφs/Mons stimulated through CCR1-CCL9 interaction. Significant functional suppression of all the three innate immune cell types occurred during the early stage of infection. In summary, we present a dynamic immune landscape, at single-cell resolution, of murine dLNs involved in the response to Y. pestis infection, which may facilitate the understanding of the plague pathogenesis of during the early stage of infection.
Collapse
|
4
|
Bekere I, Huang J, Schnapp M, Rudolph M, Berneking L, Ruckdeschel K, Grundhoff A, Günther T, Fischer N, Aepfelbacher M. Yersinia remodels epigenetic histone modifications in human macrophages. PLoS Pathog 2021; 17:e1010074. [PMID: 34793580 PMCID: PMC8639070 DOI: 10.1371/journal.ppat.1010074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 10/28/2021] [Indexed: 01/10/2023] Open
Abstract
Various pathogens systematically reprogram gene expression in macrophages, but the underlying mechanisms are largely unknown. We investigated whether the enteropathogen Yersinia enterocolitica alters chromatin states to reprogram gene expression in primary human macrophages. Genome-wide chromatin immunoprecipitation (ChIP) seq analyses showed that pathogen-associated molecular patterns (PAMPs) induced up- or down-regulation of histone modifications (HMod) at approximately 14500 loci in promoters and enhancers. Effectors of Y. enterocolitica reorganized about half of these dynamic HMod, with the effector YopP being responsible for about half of these modulatory activities. The reorganized HMod were associated with genes involved in immune response and metabolism. Remarkably, the altered HMod also associated with 61% of all 534 known Rho GTPase pathway genes, revealing a new level in Rho GTPase regulation and a new aspect of bacterial pathogenicity. Changes in HMod were associated to varying degrees with corresponding gene expression, e. g. depending on chromatin localization and cooperation of the HMod. In summary, infection with Y. enterocolitica remodels HMod in human macrophages to modulate key gene expression programs of the innate immune response. Human pathogenic bacteria can affect epigenetic histone modifications to modulate gene expression in host cells. However, a systems biology analysis of this bacterial virulence mechanism in immune cells has not been performed. Here we analyzed genome-wide epigenetic histone modifications and associated gene expression changes in primary human macrophages infected with enteropathogenic Yersinia enterocolitica. We demonstrate that Yersinia virulence factors extensively modulate histone modifications and associated gene expression triggered by the pathogen-associated molecular patterns (PAMPs) of the bacteria. The epigenetically modulated genes are involved in several key pathways of the macrophage immune response, including the Rho GTPase pathway, revealing a novel level of Rho GTPase regulation by a bacterial pathogen. Overall, our findings provide an in-depth view of epigenetic and gene expression changes during host-pathogen interaction and might have further implications for understanding of the innate immune memory in macrophages.
Collapse
Affiliation(s)
- Indra Bekere
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- * E-mail: (IB); (MA)
| | - Jiabin Huang
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Marie Schnapp
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maren Rudolph
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Laura Berneking
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Klaus Ruckdeschel
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Adam Grundhoff
- Heinrich-Pette-Institute (HPI), Leibniz Institute for Experimental Virology, Research Group Virus Genomics, Hamburg, Germany
| | - Thomas Günther
- Heinrich-Pette-Institute (HPI), Leibniz Institute for Experimental Virology, Research Group Virus Genomics, Hamburg, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- * E-mail: (IB); (MA)
| |
Collapse
|
5
|
Price SL, Vadyvaloo V, DeMarco JK, Brady A, Gray PA, Kehl-Fie TE, Garneau-Tsodikova S, Perry RD, Lawrenz MB. Yersiniabactin contributes to overcoming zinc restriction during Yersinia pestis infection of mammalian and insect hosts. Proc Natl Acad Sci U S A 2021; 118:e2104073118. [PMID: 34716262 PMCID: PMC8612365 DOI: 10.1073/pnas.2104073118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/09/2021] [Indexed: 02/04/2023] Open
Abstract
Yersinia pestis causes human plague and colonizes both a mammalian host and a flea vector during its transmission cycle. A key barrier to bacterial infection is the host's ability to actively sequester key biometals (e.g., iron, zinc, and manganese) required for bacterial growth. This is referred to as nutritional immunity. Mechanisms to overcome nutritional immunity are essential virulence factors for bacterial pathogens. Y. pestis produces an iron-scavenging siderophore called yersiniabactin (Ybt) that is required to overcome iron-mediated nutritional immunity and cause lethal infection. Recently, Ybt has been shown to bind to zinc, and in the absence of the zinc transporter ZnuABC, Ybt improves Y. pestis growth in zinc-limited medium. These data suggest that, in addition to iron acquisition, Ybt may also contribute to overcoming zinc-mediated nutritional immunity. To test this hypothesis, we used a mouse model defective in iron-mediated nutritional immunity to demonstrate that Ybt contributes to virulence in an iron-independent manner. Furthermore, using a combination of bacterial mutants and mice defective in zinc-mediated nutritional immunity, we identified calprotectin as the primary barrier for Y. pestis to acquire zinc during infection and that Y. pestis uses Ybt to compete with calprotectin for zinc. Finally, we discovered that Y. pestis encounters zinc limitation within the flea midgut, and Ybt contributes to overcoming this limitation. Together, these results demonstrate that Ybt is a bona fide zinc acquisition mechanism used by Y. pestis to surmount zinc limitation during the infection of both the mammalian and insect hosts.
Collapse
Affiliation(s)
- Sarah L Price
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Viveka Vadyvaloo
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA 99164
| | - Jennifer K DeMarco
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40292
| | - Amanda Brady
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Phoenix A Gray
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202
| | - Thomas E Kehl-Fie
- Department of Microbiology and Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Champaign, IL 61820
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY 40536
| | - Robert D Perry
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky School of Medicine, Lexington, KY 40506
| | - Matthew B Lawrenz
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202;
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, KY 40292
| |
Collapse
|
6
|
Kilgore PB, Sha J, Andersson JA, Motin VL, Chopra AK. A new generation needle- and adjuvant-free trivalent plague vaccine utilizing adenovirus-5 nanoparticle platform. NPJ Vaccines 2021; 6:21. [PMID: 33514747 PMCID: PMC7846801 DOI: 10.1038/s41541-020-00275-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
A plague vaccine with a fusion cassette of YscF, F1, and LcrV encoding genes in an adenovirus-5 vector (rAd5-YFV) is evaluated for efficacy and immune responses in mice. Two doses of the vaccine provides 100% protection when administered intranasally against challenge with Yersinia pestis CO92 or its isogenic F1 mutant in short- or long- term immunization in pneumonic/bubonic plague models. The corresponding protection rates drop in rAd5-LcrV monovalent vaccinated mice in plague models. The rAd5-YFV vaccine induces superior humoral, mucosal and cell-mediated immunity, with clearance of the pathogen. Immunization of mice with rAd5-YFV followed by CO92 infection dampens proinflammatory cytokines and neutrophil chemoattractant production, while increasing Th1- and Th2-cytokine responses as well as macrophage/monocyte chemo-attractants when compared to the challenge control animals. This is a first study showing complete protection of mice from pneumonic/bubonic plague with a viral vector-based vaccine without the use of needles and the adjuvant.
Collapse
Affiliation(s)
- Paul B. Kilgore
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA
| | - Jian Sha
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jourdan A. Andersson
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA
| | - Vladimir L. Motin
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Department of Pathology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX USA
| | - Ashok K. Chopra
- grid.176731.50000 0001 1547 9964Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX USA ,grid.176731.50000 0001 1547 9964Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX USA
| |
Collapse
|
7
|
Transcriptomic profiling of the digestive tract of the rat flea, Xenopsylla cheopis, following blood feeding and infection with Yersinia pestis. PLoS Negl Trop Dis 2020; 14:e0008688. [PMID: 32946437 PMCID: PMC7526888 DOI: 10.1371/journal.pntd.0008688] [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: 02/04/2020] [Revised: 09/30/2020] [Accepted: 08/10/2020] [Indexed: 01/29/2023] Open
Abstract
Yersinia pestis, the causative agent of plague, is a highly lethal pathogen transmitted by the bite of infected fleas. Once ingested by a flea, Y. pestis establish a replicative niche in the gut and produce a biofilm that promotes foregut colonization and transmission. The rat flea Xenopsylla cheopis is an important vector to several zoonotic bacterial pathogens including Y. pestis. Some fleas naturally clear themselves of infection; however, the physiological and immunological mechanisms by which this occurs are largely uncharacterized. To address this, RNA was extracted, sequenced, and distinct transcript profiles were assembled de novo from X. cheopis digestive tracts isolated from fleas that were either: 1) not fed for 5 days; 2) fed sterile blood; or 3) fed blood containing ~5x108 CFU/ml Y. pestis KIM6+. Analysis and comparison of the transcript profiles resulted in identification of 23 annotated (and 11 unknown or uncharacterized) digestive tract transcripts that comprise the early transcriptional response of the rat flea gut to infection with Y. pestis. The data indicate that production of antimicrobial peptides regulated by the immune-deficiency pathway (IMD) is the primary flea immune response to infection with Y. pestis. The remaining infection-responsive transcripts, not obviously associated with the immune response, were involved in at least one of 3 physiological themes: 1) alterations to chemosensation and gut peristalsis; 2) modification of digestion and metabolism; and 3) production of chitin-binding proteins (peritrophins). Despite producing several peritrophin transcripts shortly after feeding, including a subset that were infection-responsive, no thick peritrophic membrane was detectable by histochemistry or electron microscopy of rat flea guts for the first 24 hours following blood-feeding. Here we discuss the physiological implications of rat flea infection-responsive transcripts, the function of X. cheopis peritrophins, and the mechanisms by which Y. pestis may be cleared from the flea gut. The goal of this study was to characterize the transcriptional response of the digestive tract of the rat flea, Xenopsylla cheopis, to infection with Yersinia pestis, the causative agent of plague. This flea is generally considered the most prevalent and efficient vector of Y. pestis. Because most pathogens transmitted by fleas, including Y. pestis, reside in the insect digestive tract prior to transmission, the transcriptional program induced in the gut epithelium likely influences bacterial colonization of the flea. To determine the specific response to infection, RNA profiles were generated from fleas that were either unfed, fed sterile blood, or fed blood containing Y. pestis. Comparative analyses of the transcriptomes resulted in identification of 34 infection-responsive transcripts. The functions of these differentially regulated genes indicate that infection of fleas with Y. pestis induces a limited immune response and potentially alters the insect’s behavior, metabolism, and other aspects of its physiology. Based on these data, we describe potential mechanisms fleas use to eliminate bacteria and the corresponding strategies Y. pestis uses to resist elimination. These findings may be helpful for developing targeted strategies to make fleas resistant to microbial infection and thereby reduce the incidence of diseases they spread.
Collapse
|
8
|
Liu B, Bai L, Fu Y, Zhao S, Liu H, Wang R, Wang W, Li Y, Tao Y, Wang Z, Fan J, Liu E. Genetic and molecular features for hepadnavirus and plague infections in the Himalayan marmot. Genome 2020; 63:307-317. [PMID: 32308030 DOI: 10.1139/gen-2019-0161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The Himalayan marmot (Marmota himalayana), a natural host and transmitter of plague, is also susceptible to the hepadnavirus infection. To reveal the genetic basis of the hepadnavirus susceptibility and the immune response to plague, we systematically characterized the features of immune genes in Himalayan marmot with those of human and mouse. We found that the entire major histocompatibility complex region and the hepatitis B virus pathway genes of the Himalayan marmot were conserved with those of humans. A Trim (tripartite motif) gene cluster involved in immune response and antiviral activity displays dynamic evolution, which is reflected by the duplication of Trim5 and the absence of Trim22 and Trim34. Three key regions of Ntcp, which is critical for hepatitis B virus entry, had high identity among seven species of Marmota. Moreover, we observed a severe alveolar hemorrhage, inflammatory infiltrate in the infected lungs and livers from Himalayan marmots after infection of EV76, a live attenuated Yersinia pestis strain. Lots of immune genes were remarkably up-regulated, which several hub genes Il2rγ, Tra29, and Nlrp7 are placed at the center of the gene network. These findings suggest that Himalayan marmot is a potential animal model for study on the hepadnavirus and plague infection.
Collapse
Affiliation(s)
- Baoning Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Yu Fu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Haiqing Liu
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, Qinghai 811602, China
| | - Rong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Yandong Li
- Department of Pathology, First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710000, China
| | - Yuanqing Tao
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, Qinghai 811602, China
| | - Zhongdong Wang
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, Qinghai 811602, China
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| |
Collapse
|
9
|
Liu B, Bai L, Yu Q, Hu F, Wu J, Zhao S, Wang R, Wang W, Tao Y, Fan J, Liu E. iMarmot: an integrative platform for comparative and functional genomics of marmots. BMC Genomics 2020; 21:266. [PMID: 32228437 PMCID: PMC7104516 DOI: 10.1186/s12864-020-6697-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background Marmots are large Holarctic rodents with unique biological features, making them potential animal models in various research fields. Due to the rapid accumulation of the genetic data in marmots, a highly integrative database is urgent needed. Description iMarmot is freely available on the web at http://www.marmotdb.org/ and currently contains the biological information of 14 marmots, genomic sequence of 6 marmots, syntenic relationship and orthologs among 3 marmots, and expression profiles of several hibernators and plague hosts. To assist with the genomic and transcriptomic analysis, we also integrated a set of analysis and visualization tools, such as KEGG or GO enrichment analysis, PCA, Blast, Muscle, GeneWise, Lastz, and JBrowse. Particularly, one DEGs (differentially expressed genes) module has been implemented in this database to visualize the gene expression changes in hibernators and plague hosts. Conclusion This database will provide comprehensive information and analysis platform for researchers interested in understanding the biological features of marmots.
Collapse
Affiliation(s)
- Baoning Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Qingqing Yu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Fang Hu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Jing Wu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Laboratory Animal Center, Ningxia Medical University, Yinchuan, 750004, Gansu, China
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Rong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China.,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China
| | - Yuanqing Tao
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, 811602, Qinghai, China
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, No.76, Yanta West Road, Xi'an, 710061, Shaanxi, China. .,Research Institute of Atherosclerotic Disease, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
10
|
Redundant and Cooperative Roles for Yersinia pestis Yop Effectors in the Inhibition of Human Neutrophil Exocytic Responses Revealed by Gain-of-Function Approach. Infect Immun 2020; 88:IAI.00909-19. [PMID: 31871100 DOI: 10.1128/iai.00909-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Yersinia pestis causes a rapid, lethal disease referred to as plague. Y. pestis actively inhibits the innate immune system to generate a noninflammatory environment during early stages of infection to promote colonization. The ability of Y. pestis to create this early noninflammatory environment is in part due to the action of seven Yop effector proteins that are directly injected into host cells via a type 3 secretion system (T3SS). While each Yop effector interacts with specific host proteins to inhibit their function, several Yop effectors either target the same host protein or inhibit converging signaling pathways, leading to functional redundancy. Previous work established that Y. pestis uses the T3SS to inhibit neutrophil respiratory burst, phagocytosis, and release of inflammatory cytokines. Here, we show that Y. pestis also inhibits release of granules in a T3SS-dependent manner. Moreover, using a gain-of-function approach, we discovered previously hidden contributions of YpkA and YopJ to inhibition and that cooperative actions by multiple Yop effectors are required to effectively inhibit degranulation. Independent from degranulation, we also show that multiple Yop effectors can inhibit synthesis of leukotriene B4 (LTB4), a potent lipid mediator released by neutrophils early during infection to promote inflammation. Together, inhibition of these two arms of the neutrophil response likely contributes to the noninflammatory environment needed for Y. pestis colonization and proliferation.
Collapse
|
11
|
Nilsson P, Solbakken MH, Schmid BV, Orr RJS, Lv R, Cui Y, Song Y, Zhang Y, Baalsrud HT, Tørresen OK, Stenseth NC, Yang R, Jakobsen KS, Easterday WR, Jentoft S. The Genome of the Great Gerbil Reveals Species-Specific Duplication of an MHCII Gene. Genome Biol Evol 2020; 12:3832-3849. [PMID: 31971556 PMCID: PMC7046166 DOI: 10.1093/gbe/evaa008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
The great gerbil (Rhombomys opimus) is a social rodent living in permanent, complex burrow systems distributed throughout Central Asia, where it serves as the main host of several important vector-borne infectious pathogens including the well-known plague bacterium (Yersinia pestis). Here, we present a continuous annotated genome assembly of the great gerbil, covering over 96% of the estimated 2.47-Gb genome. Taking advantage of the recent genome assemblies of the sand rat (Psammomys obesus) and the Mongolian gerbil (Meriones unguiculatus), comparative immunogenomic analyses reveal shared gene losses within TLR gene families (i.e., TLR8, TLR10, and the entire TLR11-subfamily) for Gerbillinae, accompanied with signs of diversifying selection of TLR7 and TLR9. Most notably, we find a great gerbil-specific duplication of the MHCII DRB locus. In silico analyses suggest that the duplicated gene provides high peptide binding affinity for Yersiniae epitopes as well as Leishmania and Leptospira epitopes, putatively leading to increased capability to withstand infections by these pathogens. Our study demonstrates the power of whole-genome sequencing combined with comparative genomic analyses to gain deeper insight into the immunogenomic landscape of the great gerbil and its close relatives.
Collapse
Affiliation(s)
- Pernille Nilsson
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - Monica H Solbakken
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - Boris V Schmid
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | | | - Ruichen Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yujiang Zhang
- Xinjiang Center for Disease Control and Prevention, Urumqi, China
| | - Helle T Baalsrud
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - Ole K Tørresen
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - Nils Chr Stenseth
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
- Ministry of Education Key Laboratory for Earth System Modeling, Department of Earth System Science, Tsinghua University, Beijing, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Kjetill S Jakobsen
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - William Ryan Easterday
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| | - Sissel Jentoft
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Norway
| |
Collapse
|
12
|
Hamzabegovic F, Goll JB, Hooper WF, Frey S, Gelber CE, Abate G. Flagellin adjuvanted F1/V subunit plague vaccine induces T cell and functional antibody responses with unique gene signatures. NPJ Vaccines 2020; 5:6. [PMID: 31993217 PMCID: PMC6978331 DOI: 10.1038/s41541-020-0156-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
Yersinia pestis, the cause of plague, could be weaponized. Unfortunately, development of new vaccines is limited by lack of correlates of protection. We used pre- and post-vaccination sera and peripheral blood mononuclear cells from a flagellin adjuvanted F1/V vaccine trial to evaluate for protective markers. Here, we report for the first time in humans that inverse caspase-3 levels, which are measures of protective antibody, significantly increased by 29% and 75% on days 14 and 28 post-second vaccination, respectively. In addition, there were significant increases in T-cell responses on day 28 post-second vaccination. The strongest positive and negative correlations between protective antibody levels and gene expression signatures were identified for IFNG and ENSG00000225107 genes, respectively. Flagellin/F1/V subunit vaccine induced macrophage-protective antibody and significant CD4+ T-cell responses. Several genes associated with these responses were identified that could serve as potential correlates of protection.
Collapse
Affiliation(s)
- Fahreta Hamzabegovic
- Division of Infectious Diseases, Allergy and Immunology, Saint Louis University, Saint Louis, MO USA
| | | | | | - Sharon Frey
- Division of Infectious Diseases, Allergy and Immunology, Saint Louis University, Saint Louis, MO USA
| | | | - Getahun Abate
- Division of Infectious Diseases, Allergy and Immunology, Saint Louis University, Saint Louis, MO USA
| |
Collapse
|
13
|
Sun W, Singh AK. Plague vaccine: recent progress and prospects. NPJ Vaccines 2019; 4:11. [PMID: 30792905 PMCID: PMC6379378 DOI: 10.1038/s41541-019-0105-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/19/2018] [Indexed: 01/14/2023] Open
Abstract
Three great plague pandemics, resulting in nearly 200 million deaths in human history and usage as a biowarfare agent, have made Yersinia pestis as one of the most virulent human pathogens. In late 2017, a large plague outbreak raged in Madagascar attracted extensive attention and caused regional panics. The evolution of local outbreaks into a pandemic is a concern of the Centers for Disease Control and Prevention (CDC) in plague endemic regions. Until now, no licensed plague vaccine is available. Prophylactic vaccination counteracting this disease is certainly a primary choice for its long-term prevention. In this review, we summarize the latest advances in research and development of plague vaccines.
Collapse
Affiliation(s)
- Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208 USA
| | - Amit K. Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208 USA
| |
Collapse
|
14
|
Gautam A, Muhie S, Chakraborty N, Hoke A, Donohue D, Miller SA, Hammamieh R, Jett M. Metabolomic analyses reveal lipid abnormalities and hepatic dysfunction in non-human primate model for Yersinia pestis. Metabolomics 2018; 15:2. [PMID: 30830480 PMCID: PMC6311182 DOI: 10.1007/s11306-018-1457-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Pneumonic plague is caused by the aerosolized form of Yersinia pestis and is a highly virulent infection with complex clinical consequences, and without treatment, the fatality rate approaches 100%. The exact mechanisms of disease progression are unclear, with limited work done using metabolite profiling to study disease progression. OBJECTIVE The aim of this pilot study was to profile the plasma metabolomics in an animal model of Y. pestis infection. METHODS In this study, African Green monkeys were challenged with the highly virulent, aerosolized Y. pestis strain CO92, and untargeted metabolomics profiling of plasma was performed using liquid and gas chromatography with mass spectrometry. RESULTS At early time points post-exposure, we found significant increases in polyunsaturated, long chain fatty acid metabolites with p values ranging from as low as 0.000001 (ratio = 1.94) for the metabolite eicosapentaenoate to 0.04 (ratio = 1.36) for the metabolite adrenate when compared to time-matched controls. Multiple acyl carnitines metabolites were increased at earlier time points and could be a result of fatty acid oxidation defects with p values ranging from as low as 0.00001 (ratio = 2.95) for the metabolite octanoylcarnitine to 0.04 (ratio = 1.33) for metabolite deoxycarnitine when compared to time-matched controls. Dicarboxylic acids are important metabolic products of fatty acids oxidation, and when compared to time matched controls, were higher at earlier time points where metabolite tetradecanedioate has a ratio of 4.09 with significant p value of 0.000002 and adipate with a ratio of 1.12 and p value of 0.004. The metabolites from lysolipids (with significant p values ranging from 0.00006 for 1-oleoylglycerophosphoethanolamine to 0.04 for 1-stearoylglycerophosphoethanolamine and a ratio of 0.47 and 0.78, respectively) and bile acid metabolism (with significant p values ranging from 0.02 for cholate to 0.04 for deoxycholate and a ratio of 0.39 and 0.66, respectively) pathways were significantly lower compared to their time-matched controls during the entire course of infection. Metabolite levels from amino acid pathways were disrupted, and a few from the leucine, isoleucine and valine pathway were significantly higher (p values ranging from 0.002 to 0.04 and ratios ranging from 1.3 to 1.5, respectively), whereas metabolites from the urea cycle, arginine and proline pathways were significantly lower (p values ranging from 0.00008 to 0.02 and ratios ranging from 0.5 to 0.7, respectively) during the course of infection. CONCLUSIONS The involvement of several lipid pathways post-infection suggested activation of pathways linked to inflammation and oxidative stress. Metabolite data further showed increased energy demand, and multiple metabolites indicated potential hepatic dysfunction. Integration of blood metabolomics and transcriptomics data identified linoleate as a core metabolite with cross-talk with multiple genes from various time points. Collectively, the data from this study provided new insights into the mechanisms of Y. pestis pathogenesis that may aid in development of therapeutics.
Collapse
Affiliation(s)
- Aarti Gautam
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
| | - Seid Muhie
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
- The Geneva Foundation, Fort Detrick, MD, USA
| | - Nabarun Chakraborty
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
- The Geneva Foundation, Fort Detrick, MD, USA
| | - Allison Hoke
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
- The Geneva Foundation, Fort Detrick, MD, USA
| | - Duncan Donohue
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
- The Geneva Foundation, Fort Detrick, MD, USA
| | - Stacy Ann Miller
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
- The Geneva Foundation, Fort Detrick, MD, USA
| | - Rasha Hammamieh
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA
| | - Marti Jett
- US Army Center for Environmental Health Research, 568 Doughten Drive, Fort Detrick, MD, 21702, USA.
| |
Collapse
|
15
|
Davis KM. All Yersinia Are Not Created Equal: Phenotypic Adaptation to Distinct Niches Within Mammalian Tissues. Front Cell Infect Microbiol 2018; 8:261. [PMID: 30128305 PMCID: PMC6088192 DOI: 10.3389/fcimb.2018.00261] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/13/2018] [Indexed: 01/30/2023] Open
Abstract
Yersinia pseudotuberculosis replicates within mammalian tissues to form clustered bacterial replication centers, called microcolonies. A subset of bacterial cells within microcolonies interact directly with host immune cells, and other subsets of bacteria only interact with other bacteria. This establishes a system where subsets of Yersinia have distinct gene expression profiles, which are driven by their unique microenvironments and cellular interactions. When this leads to alterations in virulence gene expression, small subsets of bacteria can play a critical role in supporting the replication of the bacterial population, and can drive the overall disease outcome. Based on the pathology of infections with each of the three Yersinia species that are pathogenic to humans, it is likely that this specialization of bacterial subsets occurs during all Yersiniae infections. This review will describe the pathology that occurs during infection with each of the three human pathogenic Yersinia, in terms of the structure of bacterial replication centers and the specific immune cell subsets that bacteria interact with, and will also describe the outcome these interactions have or may have on bacterial gene expression.
Collapse
Affiliation(s)
- Kimberly M Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
16
|
Tissue dual RNA-seq allows fast discovery of infection-specific functions and riboregulators shaping host-pathogen transcriptomes. Proc Natl Acad Sci U S A 2017; 114:E791-E800. [PMID: 28096329 DOI: 10.1073/pnas.1613405114] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pathogenic bacteria need to rapidly adjust their virulence and fitness program to prevent eradication by the host. So far, underlying adaptation processes that drive pathogenesis have mostly been studied in vitro, neglecting the true complexity of host-induced stimuli acting on the invading pathogen. In this study, we developed an unbiased experimental approach that allows simultaneous monitoring of genome-wide infection-linked transcriptional alterations of the host and colonizing extracellular pathogens. Using this tool for Yersinia pseudotuberculosis-infected lymphatic tissues, we revealed numerous alterations of host transcripts associated with inflammatory and acute-phase responses, coagulative activities, and transition metal ion sequestration, highlighting that the immune response is dominated by infiltrating neutrophils and elicits a mixed TH17/TH1 response. In consequence, the pathogen's response is mainly directed to prevent phagocytic attacks. Yersinia up-regulates the gene and expression dose of the antiphagocytic type III secretion system (T3SS) and induces functions counteracting neutrophil-induced ion deprivation, radical stress, and nutritional restraints. Several conserved bacterial riboregulators were identified that impacted this response. The strongest influence on virulence was found for the loss of the carbon storage regulator (Csr) system, which is shown to be essential for the up-regulation of the T3SS on host cell contact. In summary, our established approach provides a powerful tool for the discovery of infection-specific stimuli, induced host and pathogen responses, and underlying regulatory processes.
Collapse
|
17
|
Yang H, Wang T, Tian G, Zhang Q, Wu X, Xin Y, Yan Y, Tan Y, Cao S, Liu W, Cui Y, Yang R, Du Z. Host transcriptomic responses to pneumonic plague reveal that Yersinia pestis inhibits both the initial adaptive and innate immune responses in mice. Int J Med Microbiol 2017; 307:64-74. [DOI: 10.1016/j.ijmm.2016.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 11/06/2016] [Accepted: 11/10/2016] [Indexed: 01/12/2023] Open
|
18
|
Du Z, Wang X. Pathology and Pathogenesis of Yersinia pestis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 918:193-222. [DOI: 10.1007/978-94-024-0890-4_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
Zhang Y, Roth TL, Gray EE, Chen H, Rodda LB, Liang Y, Ventura P, Villeda S, Crocker PR, Cyster JG. Migratory and adhesive cues controlling innate-like lymphocyte surveillance of the pathogen-exposed surface of the lymph node. eLife 2016; 5. [PMID: 27487469 PMCID: PMC5017864 DOI: 10.7554/elife.18156] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/30/2016] [Indexed: 12/19/2022] Open
Abstract
Lymph nodes (LNs) contain innate-like lymphocytes that survey the subcapsular sinus (SCS) and associated macrophages for pathogen entry. The factors promoting this surveillance behavior have not been defined. Here, we report that IL7RhiCcr6+ lymphocytes in mouse LNs rapidly produce IL17 upon bacterial and fungal challenge. We show that these innate-like lymphocytes are mostly LN resident. Ccr6 is required for their accumulation near the SCS and for efficient IL17 induction. Migration into the SCS intrinsically requires S1pr1, whereas movement from the sinus into the parenchyma involves the integrin LFA1 and its ligand ICAM1. CD169, a sialic acid-binding lectin, helps retain the cells within the sinus, preventing their loss in lymph flow. These findings establish a role for Ccr6 in augmenting innate-like lymphocyte responses to lymph-borne pathogens, and they define requirements for cell movement between parenchyma and SCS in what we speculate is a program of immune surveillance that helps achieve LN barrier immunity. DOI:http://dx.doi.org/10.7554/eLife.18156.001 The lymphatic system is a network of vessels and a vital part of our immune system. Amongst other things, the lymphatic system carries microbes that have entered the body – for example via to a cut or mosquito bite – to small, oval-shaped organs called lymph nodes. The lymph nodes are packed with immune cells that can be activated to help fight off infections, however certain microbes actually replicate inside the lymph nodes themselves. Lymph nodes protect themselves from these infections by having some pre-armed immune cells that are ready to respond rapidly as soon as an invading microbe is detected. These cells, referred to as innate-like lymphocytes, position themselves at the exposed surfaces of the lymph node – the locations where microbes are most likely to enter the organ. However, it was not known which cues caused these immune cells to assemble and remain at these locations. Zhang et al. now reveal that a signaling molecule called CCL20 attracts the innate-like lymphocytes to the lymph node’s exposed surfaces, while a protein known as CD169 helps to securely attach the innate-like lymphocytes in place. Further experiments then confirmed that positioning the innate-like lymphocytes at this location made mice more able to fight off the disease-causing bacterium Staphyloccus aureus. Unexpectedly, Zhang et al. also found that innate-like lymphocytes can move from the surfaces of lymph node through to the underlying tissue. This unusual migratory behavior might allow the lymphocytes to search a larger area for the infectious microbes, though further studies are needed to test this hypothesis. Future studies are also likely to focus on elucidating how the innate-like lymphocytes recognize different types of invaders, and how their activity keeps the lymph nodes healthy. DOI:http://dx.doi.org/10.7554/eLife.18156.002
Collapse
Affiliation(s)
- Yang Zhang
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Theodore L Roth
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Elizabeth E Gray
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Hsin Chen
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Lauren B Rodda
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Yin Liang
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Patrick Ventura
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| | - Saul Villeda
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, United Kingdom.,College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jason G Cyster
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
20
|
Levy Y, Vagima Y, Tidhar A, Zauberman A, Aftalion M, Gur D, Fogel I, Chitlaru T, Flashner Y, Mamroud E. Adjunctive Corticosteroid Treatment Against Yersinia pestis Improves Bacterial Clearance, Immunopathology, and Survival in the Mouse Model of Bubonic Plague. J Infect Dis 2016; 214:970-7. [PMID: 27402776 DOI: 10.1093/infdis/jiw290] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/28/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Plague is initiated by Yersinia pestis, a highly virulent bacterial pathogen. In late stages of the infection, bacteria proliferate extensively in the internal organs despite the massive infiltration of neutrophils. The ineffective inflammatory response associated with tissue damage may contribute to the low efficacy of antiplague therapies during late stages of the infection. In the present study, we address the possibility of improving therapeutic efficacy by combining corticosteroid administration with antibody therapy in the mouse model of bubonic plague. METHODS Mice were subcutaneously infected with a fully virulent Y. pestis strain and treated at progressive stages of the disease with anti-Y. pestis antibodies alone or in combination with the corticosteroid methylprednisolone. RESULTS The addition of methylprednisolone to antibody therapy correlated with improved mouse survival, a significant decrease in the amount of neutrophils and matrix metalloproteinase 9 in the tissues, and the mitigation of tissue damage. Interestingly, the combined treatment led to a decrease in the bacterial loads in infected organs. CONCLUSIONS Corticosteroids induce an unexpectedly effective antibacterial response apart from their antiinflammatory properties, thereby improving treatment efficacy.
Collapse
Affiliation(s)
- Yinon Levy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Yaron Vagima
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Avital Tidhar
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Ayelet Zauberman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - David Gur
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Itay Fogel
- Surgeon General Headquarters, IDF Medical Corps, Tel Hashomer, Israel
| | - Theodor Chitlaru
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Yehuda Flashner
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| | - Emanuelle Mamroud
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona
| |
Collapse
|
21
|
Temporal Progression of Pneumonic Plague in Blood of Nonhuman Primate: A Transcriptomic Analysis. PLoS One 2016; 11:e0151788. [PMID: 27003632 PMCID: PMC4803270 DOI: 10.1371/journal.pone.0151788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 03/02/2016] [Indexed: 01/07/2023] Open
Abstract
Early identification of impending illness during widespread exposure to a pathogenic agent offers a potential means to initiate treatment during a timeframe when it would be most likely to be effective and has the potential to identify novel therapeutic strategies. The latter could be critical, especially as antibiotic resistance is becoming widespread. In order to examine pre-symptomatic illness, African green monkeys were challenged intranasally with aerosolized Yersinia pestis strain CO92 and blood samples were collected in short intervals from 45 m till 42 h post-exposure. Presenting one of the first genomic investigations of a NHP model challenged by pneumonic plague, whole genome analysis was annotated in silico and validated by qPCR assay. Transcriptomic profiles of blood showed early perturbation with the number of differentially expressed genes increasing until 24 h. By then, Y. pestis had paralyzed the host defense, as suggested by the functional analyses. Early activation of the apoptotic networks possibly facilitated the pathogen to overwhelm the defense mechanisms, despite the activation of the pro-inflammatory mechanism, toll-like receptors and microtubules at the port-of-entry. The overexpressed transcripts encoding an early pro-inflammatory response particularly manifested in active lymphocytes and ubiquitin networks were a potential deviation from the rodent models, which needs further verification. In summary, the present study recognized a pattern of Y. pestis pathogenesis potentially more applicable to the human system. Independent validation using the complementary omics approach with comprehensive evaluation of the organs, such as lungs which showed early bacterial infection, is essential.
Collapse
|
22
|
Perry RD, Bobrov AG, Fetherston JD. The role of transition metal transporters for iron, zinc, manganese, and copper in the pathogenesis of Yersinia pestis. Metallomics 2016; 7:965-78. [PMID: 25891079 DOI: 10.1039/c4mt00332b] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Yersinia pestis, the causative agent of bubonic, septicemic and pneumonic plague, encodes a multitude of Fe transport systems. Some of these are defective due to frameshift or IS element insertions, while others are functional in vitro but have no established role in causing infections. Indeed only 3 Fe transporters (Ybt, Yfe and Feo) have been shown to be important in at least one form of plague. The yersiniabactin (Ybt) system is essential in the early dermal/lymphatic stages of bubonic plague, irrelevant in the septicemic stage, and critical in pneumonic plague. Two Mn transporters have been characterized (Yfe and MntH). These two systems play a role in bubonic plague but the double yfe mntH mutant is fully virulent in a mouse model of pneumonic plague. The same in vivo phenotype occurs with a mutant lacking two (Yfe and Feo) of four ferrous transporters. A role for the Ybt siderophore in Zn acquisition has been revealed. Ybt-dependent Zn acquisition uses a transport system completely independent of the Fe-Ybt uptake system. Together Ybt components and ZnuABC play a critical role in Zn acquisition in vivo. Single mutants in either system retain high virulence in a mouse model of septicemic plague while the double mutant is completely avirulent.
Collapse
Affiliation(s)
- Robert D Perry
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA.
| | | | | |
Collapse
|
23
|
Yang R, Cui Y, Bi Y. Perspectives on Yersinia pestis: A Model for Studying Zoonotic Pathogens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 918:377-391. [PMID: 27722871 DOI: 10.1007/978-94-024-0890-4_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Yersinia pestis is a typical zoonotic bacterial pathogen. The following reasons make this pathogen a model for studying zoonotic pathogens: (1) Its unique lifestyle makes Y. pestis an ideal model for studying host-vector-environment-pathogen interactions; (2) population diversity characters in Y. pestis render it a model species for studying monomorphic bacterial evolution; (3) the pathogenic features of bacteria provide us with good opportunities to study human immune responses; (4) typical animal and vector models of Y. pestis infection create opportunities for experimental studies on pathogenesis and evolution; and (5) repeated pandemics and local outbreaks provide us with clues about the infectious disease outbreaks that have occurred in human history.
Collapse
Affiliation(s)
- Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology, No. 20, Dongdajie, Fengtai, Beijing, 100071, China.
| | - Yujun Cui
- Beijing Institute of Microbiology and Epidemiology, No. 20, Dongdajie, Fengtai, Beijing, 100071, China
| | - Yujing Bi
- Beijing Institute of Microbiology and Epidemiology, No. 20, Dongdajie, Fengtai, Beijing, 100071, China
| |
Collapse
|
24
|
Yang R, Motin VL. Yersinia pestis in the Age of Big Data. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 918:257-272. [PMID: 27722866 DOI: 10.1007/978-94-024-0890-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
Abstract
As omics-driven technologies developed rapidly, genomics, transcriptomics, proteomics, metabolomics and other omics-based data have been accumulated in unprecedented speed. Omics-driven big data in biology have changed our way of research. "Big science" has promoted our understanding of biology in a holistic overview that is impossibly achieved by traditional hypothesis-driven research. In this chapter, we gave an overview of omics-driven research on Y. pestis, provided a way of thinking on Yersinia pestis research in the age of big data, and made some suggestions to integrate omics-based data for systems understanding of Y. pestis.
Collapse
Affiliation(s)
- Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology, No. Dongdajie, Fengtai, Beijing, 100071, China.
| | - Vladimir L Motin
- Departments of Pathology and Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
25
|
Role of Tellurite Resistance Operon in Filamentous Growth of Yersinia pestis in Macrophages. PLoS One 2015; 10:e0141984. [PMID: 26536670 PMCID: PMC4633105 DOI: 10.1371/journal.pone.0141984] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/15/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Yersinia pestis initiates infection by parasitism of host macrophages. In response to macrophage infections, intracellular Y. pestis can assume a filamentous cellular morphology which may mediate resistance to host cell innate immune responses. We previously observed the expression of Y. pestis tellurite resistance proteins TerD and TerE from the terZABCDE operon during macrophage infections. Others have observed a filamentous response associated with expression of tellurite resistance operon in Escherichia coli exposed to tellurite. Therefore, in this study we examine the potential role of Y. pestis tellurite resistance operon in filamentous cellular morphology during macrophage infections. PRINCIPAL FINDINGS In vitro treatment of Y. pestis culture with sodium tellurite (Na2TeO3) caused the bacterial cells to assume a filamentous phenotype similar to the filamentous phenotype observed during macrophage infections. A deletion mutant for genes terZAB abolished the filamentous morphologic response to tellurite exposure or intracellular parasitism, but without affecting tellurite resistance. However, a terZABCDE deletion mutant abolished both filamentous morphologic response and tellurite resistance. Complementation of the terZABCDE deletion mutant with terCDE, but not terZAB, partially restored tellurite resistance. When the terZABCDE deletion mutant was complemented with terZAB or terCDE, Y. pestis exhibited filamentous morphology during macrophage infections as well as while these complemented genes were being expressed under an in vitro condition. Further in E. coli, expression of Y. pestis terZAB, but not terCDE, conferred a filamentous phenotype. CONCLUSIONS These findings support the role of Y. pestis terZAB mediation of the filamentous response phenotype; whereas, terCDE confers tellurite resistance. Although the beneficial role of filamentous morphological responses by Y. pestis during macrophage infections is yet to be fully defined, it may be a bacterial adaptive strategy to macrophage associated stresses.
Collapse
|
26
|
Spinner JL, Hasenkrug AM, Shannon JG, Kobayashi SD, Hinnebusch BJ. Role of the Yersinia YopJ protein in suppressing interleukin-8 secretion by human polymorphonuclear leukocytes. Microbes Infect 2015; 18:21-9. [PMID: 26361732 DOI: 10.1016/j.micinf.2015.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/20/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022]
Abstract
Polymorphonuclear leukocytes, in addition to their direct bactericidal activities, produce cytokines involved in the activation and regulation of the innate and adaptive immune response to infection. In this study we evaluated the cytokine response of human PMNs following incubation with the pathogenic Yersinia species. Yersinia pestis strains with the pCD1 virulence plasmid, which encodes cytotoxic Yop proteins that are translocated into host cells, stimulated little or no cytokine production compared to pCD1-negative strains. In particular, PMNs incubated with pCD1-negative Y. pestis secreted 1000-fold higher levels of interleukin-8 (IL-8 or CXCL8), a proinflammatory chemokine important for PMN recruitment and activation. Deletion of yopE, -H, -T, -M or ypkA had no effect on pCD1-dependent inhibition, whereas deletion of yopJ resulted in significantly increased IL-8 production. Like Y. pestis, the enteropathogenic Yersinia species inhibited IL-8 secretion by PMNs, and strains lacking the virulence plasmid induced high levels of IL-8. Our results show that virulence plasmid-encoded effector Yops, particularly YopJ, prevent IL-8 secretion by human PMNs. Suppression of the chemotactic IL-8 response by Y. pestis may contribute to the delayed PMN recruitment to the infected lymph node that typifies bubonic plague.
Collapse
Affiliation(s)
- Justin L Spinner
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 905 S. 4th St., Hamilton, Montana 59840, USA
| | - Aaron M Hasenkrug
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 905 S. 4th St., Hamilton, Montana 59840, USA
| | - Jeffrey G Shannon
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 905 S. 4th St., Hamilton, Montana 59840, USA
| | - Scott D Kobayashi
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 905 S. 4th St., Hamilton, Montana 59840, USA
| | - B Joseph Hinnebusch
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 905 S. 4th St., Hamilton, Montana 59840, USA.
| |
Collapse
|
27
|
Korhonen TK. Fibrinolytic and procoagulant activities of Yersinia pestis and Salmonella enterica. J Thromb Haemost 2015; 13 Suppl 1:S115-20. [PMID: 26149012 DOI: 10.1111/jth.12932] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Pla of the plague bacterium Yersinia pestis and PgtE of the enteropathogen Salmonella enterica are surface-exposed, transmembrane β-barrel proteases of the omptin family that exhibit a complex array of interactions with the hemostatic systems in vitro, and both proteases are established virulence factors. Pla favors fibrinolysis by direct activation of plasminogen, inactivation of the serpins plasminogen activator inhibitor-1 and α2-antiplasmin, inactivation of the thrombin-activable fibrinolysis inhibitor, and activation of single-chain urokinase. PgtE is structurally very similar but exhibits partially different functions and differ in expression control. PgtE proteolysis targets control aspects of fibrinolysis, and mimicry of matrix metalloproteinases enhances cell migration that should favor the intracellular spread of the bacterium. Enzymatic activity of both proteases is strongly influenced by the environment-induced variations in lipopolysaccharide that binds to the β-barrel. Both proteases cleave the tissue factor pathway inhibitor and thus also express procoagulant activity.
Collapse
Affiliation(s)
- T K Korhonen
- General Microbiology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
28
|
Sivaraman V, Pechous RD, Stasulli NM, Eichelberger KR, Miao EA, Goldman WE. Yersinia pestis activates both IL-1β and IL-1 receptor antagonist to modulate lung inflammation during pneumonic plague. PLoS Pathog 2015; 11:e1004688. [PMID: 25781467 PMCID: PMC4363893 DOI: 10.1371/journal.ppat.1004688] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/19/2015] [Indexed: 11/25/2022] Open
Abstract
Pneumonic plague is the most rapid and lethal form of Yersinia pestis infection. Increasing evidence suggests that Y. pestis employs multiple levels of innate immune evasion and/or suppression to produce an early “pre-inflammatory” phase of pulmonary infection, after which the disease is highly inflammatory in the lung and 100% fatal. In this study, we show that IL-1β/IL-18 cytokine activation occurs early after bacteria enter the lung, and this activation eventually contributes to pulmonary inflammation and pathology during the later stages of infection. However, the inflammatory effects of IL-1β/IL-1-receptor ligation are not observed during this first stage of pneumonic plague. We show that Y. pestis also activates the induction of IL-1 receptor antagonist (IL-1RA), and this activation likely contributes to the ability of Y. pestis to establish the initial pre-inflammatory phase of disease. Inhalation of respiratory droplets containing Yersinia pestis results in a rapidly developing and lethal pneumonia. Interestingly, early interactions between Y. pestis and host cells in the lung contribute to significant immune evasion, but also ultimately result in severe innate immune activation. Our results demonstrate that Y. pestis activates pro-inflammatory cytokines IL-1β and IL-18 in the lung early during infection. However, there is very little early pulmonary inflammation while Y. pestis continues to multiply in the lung compartment. We show that the host protein IL-1RA is activated concurrently with IL-1β, attenuating early immune activation by this cytokine. We propose that this allows the organism to replicate to high titers, eventually triggering a vigorous inflammatory response and facilitating aerosol transmission. Therefore, evaluating early host activation of IL-1RA by Y. pestis may provide therapeutic targets against pneumonic plague.
Collapse
Affiliation(s)
- Vijay Sivaraman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biology, North Carolina Central University, Durham, North Carolina, United States of America
| | - Roger D. Pechous
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nikolas M. Stasulli
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kara R. Eichelberger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Edward A. Miao
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William E. Goldman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
29
|
Evaluation of the murine immune response to Xenopsylla cheopis flea saliva and its effect on transmission of Yersinia pestis. PLoS Negl Trop Dis 2014; 8:e3196. [PMID: 25255317 PMCID: PMC4177749 DOI: 10.1371/journal.pntd.0003196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/18/2014] [Indexed: 01/20/2023] Open
Abstract
Background/Aims Arthropod-borne pathogens are transmitted into a unique intradermal microenvironment that includes the saliva of their vectors. Immunomodulatory factors in the saliva can enhance infectivity; however, in some cases the immune response that develops to saliva from prior uninfected bites can inhibit infectivity. Most rodent reservoirs of Yersinia pestis experience fleabites regularly, but the effect this has on the dynamics of flea-borne transmission of plague has never been investigated. We examined the innate and acquired immune response of mice to bites of Xenopsylla cheopis and its effects on Y. pestis transmission and disease progression in both naïve mice and mice chronically exposed to flea bites. Methods/Principal Findings The immune response of C57BL/6 mice to uninfected flea bites was characterized by flow cytometry, histology, and antibody detection methods. In naïve mice, flea bites induced mild inflammation with limited recruitment of neutrophils and macrophages to the bite site. Infectivity and host response in naïve mice exposed to flea bites followed immediately by intradermal injection of Y. pestis did not differ from that of mice infected with Y. pestis without prior flea feeding. With prolonged exposure, an IgG1 antibody response primarily directed to the predominant component of flea saliva, a family of 36–45 kDa phosphatase-like proteins, occurred in both laboratory mice and wild rats naturally exposed to X. cheopis, but a hypersensitivity response never developed. The incidence and progression of terminal plague following challenge by infective blocked fleas were equivalent in naïve mice and mice sensitized to flea saliva by repeated exposure to flea bites over a 10-week period. Conclusions Unlike what is observed with many other blood-feeding arthropods, the murine immune response to X. cheopis saliva is mild and continued exposure to flea bites leads more to tolerance than to hypersensitivity. The immune response to flea saliva had no detectable effect on Y. pestis transmission or plague pathogenesis in mice. The saliva of blood-feeding arthropods contains a variety of components that prevent blood clotting and interfere with the immune system of the vertebrate host. These properties have been shown to enhance or inhibit the transmission of different pathogens transmitted by arthropods. Yersinia pestis, the bacterial agent of plague, is maintained in nature by flea to rodent transmission cycles. Most rodents live in close association with fleas and are constantly being bitten by them, but the influence this has on plague transmission is unknown - previous studies used laboratory animals which have never experienced a flea bite. We found that flea bites caused a mild inflammatory response in mice, and eventually an antibody response to components of flea saliva, but did not significantly affect pathogenesis. The transmission of Y. pestis by infected fleas and the incidence rate of bubonic plague mortality were the same in mice that had been exposed to frequent uninfected flea bites and mice with no prior exposure to fleas. Therefore, in contrast to what has been shown for many other arthropod-borne disease systems, vector saliva did not enhance or inhibit Y. pestis infection in mice, regardless of the immune status of the host to flea saliva.
Collapse
|
30
|
Du Z, Yang H, Tan Y, Tian G, Zhang Q, Cui Y, Yanfeng Yan, Wu X, Chen Z, Cao S, Bi Y, Han Y, Wang X, Song Y, Yang R. Transcriptomic response to Yersinia pestis: RIG-I like receptor signaling response is detrimental to the host against plague. J Genet Genomics 2014; 41:379-96. [PMID: 25064677 DOI: 10.1016/j.jgg.2014.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/09/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023]
Abstract
Bacterial pathogens have evolved various mechanisms to modulate host immune responses for successful infection. In this study, RNA-sequencing technology was used to analyze the responses of human monocytes THP1 to Yersinia pestis infection. Over 6000 genes were differentially expressed over the 12 h infection. Kinetic responses of pathogen recognition receptor signaling pathways, apoptosis, antigen processing, and presentation pathway and coagulation system were analyzed in detail. Among them, RIG-I-like receptor (RLR) signaling pathway, which was established for antiviral defense, was significantly affected. Mice lacking MAVS, the adaptor of the RLR signaling pathway, were less sensitive to infection and exhibited lower IFN-β production, higher Th1-type cytokines IFN-γ and IL-12 production, and lower Th2-type cytokines IL-4 and IL-13 production in the serum compared with wild-type mice. Moreover, infection of pathogenic bacteria other than Y. pestis also altered the expression of the RLR pathway, suggesting that the response of RLR pathway to bacterial infection is a universal mechanism.
Collapse
Affiliation(s)
- Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Huiying Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yafang Tan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Guang Tian
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qingwen Zhang
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province, Xining 811602, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yanfeng Yan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaohong Wu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | - Shiyang Cao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yanping Han
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaoyi Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|
31
|
Reboul A, Lemaître N, Titecat M, Merchez M, Deloison G, Ricard I, Pradel E, Marceau M, Sebbane F. Yersinia pestis requires the 2-component regulatory system OmpR-EnvZ to resist innate immunity during the early and late stages of plague. J Infect Dis 2014; 210:1367-75. [PMID: 24813471 DOI: 10.1093/infdis/jiu274] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Plague is transmitted by fleas or contaminated aerosols. To successfully produce disease, the causal agent (Yersinia pestis) must rapidly sense and respond to rapid variations in its environment. Here, we investigated the role of 2-component regulatory systems (2CSs) in plague because the latter are known to be key players in bacterial adaptation to environmental change. Along with the previously studied PhoP-PhoQ system, OmpR-EnvZ was the only one of Y. pestis' 23 other 2CSs required for production of bubonic, septicemic, and pneumonic plague. In vitro, OmpR-EnvZ was needed to counter serum complement and leukocytes but was not required for the secretion of antiphagocyte exotoxins. In vivo, Y. pestis lacking OmpR-EnvZ did not induce an early immune response in the skin and was fully virulent in neutropenic mice. We conclude that, throughout the course of Y. pestis infection, OmpR-EnvZ is required to counter toxic effectors secreted by polymorphonuclear leukocytes in the tissues.
Collapse
Affiliation(s)
- Angéline Reboul
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Nadine Lemaître
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille Centre Hospitalier Régional Universitaire de Lille, France
| | - Marie Titecat
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille Centre Hospitalier Régional Universitaire de Lille, France
| | - Maud Merchez
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Gaspard Deloison
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Isabelle Ricard
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Elizabeth Pradel
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Michaël Marceau
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| | - Florent Sebbane
- Plague and Yersinia pestis Group, INSERM U1019 Centre National de la Recherche Scientifique UMR8204 Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille Université Lille Nord de France Centre d'Infection et d'Immunité de Lille, Université du Droit et de la Santé de Lille
| |
Collapse
|
32
|
Pradel E, Lemaître N, Merchez M, Ricard I, Reboul A, Dewitte A, Sebbane F. New insights into how Yersinia pestis adapts to its mammalian host during bubonic plague. PLoS Pathog 2014; 10:e1004029. [PMID: 24675805 PMCID: PMC3968184 DOI: 10.1371/journal.ppat.1004029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/11/2014] [Indexed: 12/22/2022] Open
Abstract
Bubonic plague (a fatal, flea-transmitted disease) remains an international public health concern. Although our understanding of the pathogenesis of bubonic plague has improved significantly over the last few decades, researchers have still not been able to define the complete set of Y. pestis genes needed for disease or to characterize the mechanisms that enable infection. Here, we generated a library of Y. pestis mutants, each lacking one or more of the genes previously identified as being up-regulated in vivo. We then screened the library for attenuated virulence in rodent models of bubonic plague. Importantly, we tested mutants both individually and using a novel, “per-pool” screening method that we have developed. Our data showed that in addition to genes involved in physiological adaption and resistance to the stress generated by the host, several previously uncharacterized genes are required for virulence. One of these genes (ympt1.66c, which encodes a putative helicase) has been acquired by horizontal gene transfer. Deletion of ympt1.66c reduced Y. pestis' ability to spread to the lymph nodes draining the dermal inoculation site – probably because loss of this gene decreased the bacteria's ability to survive inside macrophages. Our results suggest that (i) intracellular survival during the early stage of infection is important for plague and (ii) horizontal gene transfer was crucial in the acquisition of this ability. In order to understand and combat infectious diseases, it is essential to characterize the full set of genes required by pathogenic bacteria to overcome the many immunological and physiological challenges encountered during infection. Here, we used a genome-scale approach to identify genes required by the bacterium Yersinia pestis in the production of bubonic plague (a fatal, flea-borne zoonosis). Our results suggest that when colonizing the mammalian host, the bacterium (i) relies on carbohydrates as its carbon source, (ii) shifts to anaerobic respiration or fermentation and (iii) experiences and resists several (but not all) types of stress generated by the host's innate immune system. Strikingly, only a small set of genes (including horizontally acquired and uncharacterized sequences) are required for these infectious processes. Further investigations of the ypmt1,66c gene provided evidence to suggest that accretion of genetic material via horizontal transfer has played a key role in Yersinia pestis' ability to successfully initiate infection after the dermal fleabite. Lastly, we believe that (i) application of our approach to other pathogens and (ii) additional studies of selected Yersinia pestis genes important for plague pathogenesis (some of which are shared with other pathogens) will provide a better understanding of bacterial pathogenesis in general.
Collapse
Affiliation(s)
- Elizabeth Pradel
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Nadine Lemaître
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
- CHU Lille, Lille, France
| | - Maud Merchez
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Isabelle Ricard
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Angéline Reboul
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Amélie Dewitte
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Florent Sebbane
- Equipe Peste et Yersinia pestis; INSERM U1019, Lille, France
- Centre National de la Recherche Scientifique UMR8204, Lille, France
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Univ Lille Nord de France, Lille, France
- UDSL, Centre d'Infection et d'Immunité de Lille, Lille, France
- * E-mail:
| |
Collapse
|
33
|
Bi Y, Zhou J, Yang H, Wang X, Zhang X, Wang Q, Wu X, Han Y, Song Y, Tan Y, Du Z, Yang H, Zhou D, Cui Y, Zhou L, Yan Y, Zhang P, Guo Z, Wang X, Liu G, Yang R. IL-17A produced by neutrophils protects against pneumonic plague through orchestrating IFN-γ-activated macrophage programming. THE JOURNAL OF IMMUNOLOGY 2013; 192:704-13. [PMID: 24337746 DOI: 10.4049/jimmunol.1301687] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Innate immune cells, including neutrophils and macrophages, are critically involved in host antimicrobial defense responses. Intrinsic regulatory mechanisms controlling neutrophil and macrophage activities are poorly defined. In this study, we found that IL-17A, a natural signal factor, could provide protection against early pneumonic plague inflammation by coordinating the functions of neutrophils and programming of macrophages. The IL-17A level is promptly increased during the initial infection. Importantly, abrogation of IL-17A or IL-17AR significantly aggravated the infection, but mIL-17A treatment could significantly alleviate inflammatory injury, revealing that IL-17A is a critical requirement for early protection of infection. We also demonstrated that IL-17A was predominantly produced by CD11b(+)Ly6G(+) neutrophils. Although IL-17A could not significantly affect the antimicrobial responses of neutrophils, it could target the proinflammatory macrophage (M1) programming and potentiate the M1's defense against pneumonic plague. Mechanistically, IFN-γ treatment or IFN-γ-activated M1 macrophage transfer could significantly mitigate the aggravated infection of IL-17A(-/-) mice. Finally, we showed that IL-17A and IFN-γ could synergistically promote macrophage anti-infection immunity. Thus, our findings identify a previously unrecognized function of IL-17A as an intrinsic regulator in coordinating neutrophil and macrophage antimicrobial activity to provide protection against acute pneumonic plague.
Collapse
Affiliation(s)
- Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Spinner JL, Winfree S, Starr T, Shannon JG, Nair V, Steele-Mortimer O, Hinnebusch BJ. Yersinia pestis survival and replication within human neutrophil phagosomes and uptake of infected neutrophils by macrophages. J Leukoc Biol 2013; 95:389-98. [PMID: 24227798 DOI: 10.1189/jlb.1112551] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Yersinia pestis, the bacterial agent of plague, is transmitted by fleas. The bite of an infected flea deposits Y. pestis into the dermis and triggers recruitment of innate immune cells, including phagocytic PMNs. Y. pestis can subvert this PMN response and survive at the flea-bite site, disseminate, and persist in the host. Although its genome encodes a number of antiphagocytic virulence factors, phagocytosis of Y. pestis by PMNs has been observed. This study tests the hypotheses that Y. pestis, grown at the ambient temperature of the flea vector (21°C), where the major antiphagocytic virulence factors are not produced, can survive and replicate within human PMNs and can use PMNs as a route to infect macrophages subsequently. We show that Y. pestis is localized within PMN phagosomes, predominately as individual bacteria, and that intracellular bacteria can survive and replicate. Within 12 h of infection, ~70% of infected PMNs had PS on their surface and were plausibly competent for efferocytosis. With the use of live cell confocal imaging, we show that autologous HMDMs recognize and internalize infected PMNs and that Y. pestis survives and replicates within these HMDMs following efferocytosis. Addition of HMDMs to infected PMNs resulted in decreased secretion of inflammatory cytokines (compared with HMDMs incubated directly with pCD1(-) Y. pestis) and increased secretion of the anti-inflammatory cytokine IL-1ra. Thus, Y. pestis can survive and replicate within PMNs, and infected PMNs may be a route for noninflammatory infection of macrophages.
Collapse
Affiliation(s)
- Justin L Spinner
- 1.Rocky Mountain Laboratories, NIAID, NIH, 903 South 4th St., Hamilton, MT 59840, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Butler T. Plague gives surprises in the first decade of the 21st century in the United States and worldwide. Am J Trop Med Hyg 2013; 89:788-93. [PMID: 24043686 DOI: 10.4269/ajtmh.13-0191] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Plague is an ancient disease caused by the bacterium Yersinia pestis and transmitted by rodent flea bites that continues to surprise us with first-ever events. This review documents plague in human cases in the 1st decade of the 21st century and updates our knowledge of clinical manifestations, transmission during outbreaks, diagnostic testing, antimicrobial treatment, and vaccine development. In the United States, 57 persons were reported to have the disease, of which seven died. Worldwide, 21,725 persons were affected with 1,612 deaths, for a case-fatality rate of 7.4%. The Congo reported more cases than any other country, including two large outbreaks of pneumonic plague, surpassing Madagascar, which had the most cases in the previous decade. Two United States scientists suffered fatal accidental exposures: a wildlife biologist, who carried out an autopsy on a mountain lion in Arizona in 2007, and a geneticist with subclinical hemochromatosis in Chicago, who was handling an avirulent strain of Y. pestis in 2009. Antimicrobial drugs given early after the onset of symptoms prevented many deaths; those recommended for treatment and prophylaxis included gentamicin, doxycycline, and fluoroquinolones, although fluoroquinolones have not been adequately tested in humans. Fleas that do not have their guts blocked by clotted blood meals were shown to be better transmitters of plague than blocked fleas. Under development for protection against bioterrorist use, a subunit vaccine containing F1 and V antigens of Y. pestis was administered to human volunteers eliciting antibodies without any serious side effects. These events, although showing progress, suggest that plague will persist in rodent reservoirs mostly in African countries burdened by poverty and civil unrest, causing death when patients fail to receive prompt antimicrobial treatment.
Collapse
Affiliation(s)
- Thomas Butler
- Ross University School of Medicine, Portsmouth, Dominica, West Indies
| |
Collapse
|
36
|
Abstract
The majority of human Yersinia pestis infections result from introduction of bacteria into the skin by the bite of an infected flea. Once in the dermis, Y. pestis can evade the host’s innate immune response and subsequently disseminate to the draining lymph node (dLN). There, the pathogen replicates to large numbers, causing the pathognomonic bubo of bubonic plague. In this study, several cytometric and microscopic techniques were used to characterize the early host response to intradermal (i.d.) Y. pestis infection. Mice were infected i.d. with fully virulent or attenuated strains of dsRed-expressing Y. pestis, and tissues were analyzed by flow cytometry. By 4 h postinfection, there were large numbers of neutrophils in the infected dermis and the majority of cell-associated bacteria were associated with neutrophils. We observed a significant effect of the virulence plasmid (pCD1) on bacterial survival and neutrophil activation in the dermis. Intravital microscopy of i.d. Y. pestis infection revealed dynamic interactions between recruited neutrophils and bacteria. In contrast, very few bacteria interacted with dendritic cells (DCs), indicating that this cell type may not play a major role early in Y. pestis infection. Experiments using neutrophil depletion and a CCR7 knockout mouse suggest that dissemination of Y. pestis from the dermis to the dLN is not dependent on neutrophils or DCs. Taken together, the results of this study show a very rapid, robust neutrophil response to Y. pestis in the dermis and that the virulence plasmid pCD1 is important for the evasion of this response. Yersinia pestis remains a public health concern today because of sporadic plague outbreaks that occur throughout the world and the potential for its illegitimate use as a bioterrorism weapon. Since bubonic plague pathogenesis is initiated by the introduction of Y. pestis into the skin, we sought to characterize the response of the host’s innate immune cells to bacteria early after intradermal infection. We found that neutrophils, innate immune cells that engulf and destroy microbes, are rapidly recruited to the injection site, irrespective of strain virulence, indicating that Y. pestis is unable to subvert neutrophil recruitment to the site of infection. However, we saw a decreased activation of neutrophils that were associated with Y. pestis strains harboring the pCD1 plasmid, which is essential for virulence. These findings indicate a role for pCD1-encoded factors in suppressing the activation/stimulation of these cells in vivo.
Collapse
|
37
|
Korhonen TK, Haiko J, Laakkonen L, Järvinen HM, Westerlund-Wikström B. Fibrinolytic and coagulative activities of Yersinia pestis. Front Cell Infect Microbiol 2013; 3:35. [PMID: 23898467 PMCID: PMC3724046 DOI: 10.3389/fcimb.2013.00035] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 07/09/2013] [Indexed: 12/18/2022] Open
Abstract
The outer membrane protease Pla belongs to the omptin protease family spread by horizontal gene transfer into Gram-negative bacteria that infect animals or plants. Pla has adapted to support the life style of the plague bacterium Yersinia pestis. Pla has a β-barrel fold with 10 membrane-spanning β strands and five surface loops, and the barrel surface contains bound lipopolysaccharide (LPS) that is critical for the conformation and the activity of Pla. The biological activity of Pla is influenced by the structure of the surface loops around the active site groove and by temperature-induced LPS modifications. Several of the putative virulence-related functions documented for Pla in vitro address control of the human hemostatic system, i.e., coagulation and fibrinolysis. Pla activates human plasminogen to the serine protease plasmin and activates the physiological plasminogen activator urokinase. Pla also inactivates the protease inhibitors alpha-2-antiplasmin and plasminogen activator inhibitor 1 (PAI-1) and prevents the activation of thrombin-activatable fibrinolysis inhibitor (TAFI). These functions enhance uncontrolled fibrinolysis which is thought to improve Y. pestis dissemination and survival in the mammalian host, and lowered fibrin(ogen) deposition has indeed been observed in mice infected with Pla-positive Y. pestis. However, Pla also inactivates an anticoagulant, the tissue factor (TF) pathway inhibitor, which should increase fibrin formation and clotting. Thus, Pla and Y. pestis have complex interactions with the hemostatic system. Y. pestis modifies its LPS upon transfer to the mammalian host and we hypothesize that the contrasting biological activities of Pla in coagulation and fibrinolysis are influenced by LPS changes during infection.
Collapse
Affiliation(s)
- Timo K Korhonen
- General Microbiology, Department of Biosciences, University of Helsinki Helsinki, Finland.
| | | | | | | | | |
Collapse
|
38
|
Barh D, Gupta K, Jain N, Khatri G, León-Sicairos N, Canizalez-Roman A, Tiwari S, Verma A, Rahangdale S, Shah Hassan S, Rodrigues dos Santos A, Ali A, Carlos Guimarães L, Thiago Jucá Ramos R, Devarapalli P, Barve N, Bakhtiar M, Kumavath R, Ghosh P, Miyoshi A, Silva A, Kumar A, Narayan Misra A, Blum K, Baumbach J, Azevedo V. Conserved host–pathogen PPIs Globally conserved inter-species bacterial PPIs based conserved host-pathogen interactome derived novel target inC. pseudotuberculosis,C. diphtheriae,M. tuberculosis,C. ulcerans,Y. pestis, andE. colitargeted byPiper betelcompounds. Integr Biol (Camb) 2013; 5:495-509. [DOI: 10.1039/c2ib20206a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- Department of Biosciences and Biotechnology, School of Biotechnology, Fakir Mohan University, Jnan Bigyan Vihar, Balasore, Orissa, India
| | - Krishnakant Gupta
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Neha Jain
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
| | - Gourav Khatri
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Nidia León-Sicairos
- Unidad de investigacion, Facultad de Medicina, Universidad Autónoma de Sinaloa. Cedros y Sauces, Fraccionamiento Fresnos, Culiacán Sinaloa 80246, México
| | - Adrian Canizalez-Roman
- Unidad de investigacion, Facultad de Medicina, Universidad Autónoma de Sinaloa. Cedros y Sauces, Fraccionamiento Fresnos, Culiacán Sinaloa 80246, México
| | - Sandeep Tiwari
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
| | - Ankit Verma
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Sachin Rahangdale
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Syed Shah Hassan
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Amjad Ali
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luis Carlos Guimarães
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Pratap Devarapalli
- Department of Genomic Science, School of Biological Sciences, Riverside Transit Campus, Central University of Kerala, Kasaragod, India
| | - Neha Barve
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Marriam Bakhtiar
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Riverside Transit Campus, Central University of Kerala, Kasaragod, India
| | - Preetam Ghosh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- Department of Computer Science and Center for the Study of Biological Complexity, Virginia Commonwealth University, 401 West Main Street, Room E4234, P.O. Box 843019, Richmond, Virginia 23284-3019, USA
| | - Anderson Miyoshi
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Artur Silva
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil
| | - Anil Kumar
- School of Biotechnology, Devi Ahilya University, Khandwa Road Campus, Indore, MP, India
| | - Amarendra Narayan Misra
- Department of Biosciences and Biotechnology, School of Biotechnology, Fakir Mohan University, Jnan Bigyan Vihar, Balasore, Orissa, India
- Center for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ranchi, Jharkhand State, India
| | - Kenneth Blum
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal-721172, India. Fax: +91-944 955 0032; Tel: +91-944 955 0032
- University of Florida, College of Medicine, Gainesville, Florida, USA
- Global Integrated Services Unit University of Vermont Center for Clinical & Translational Science, College of Medicine, Burlington, VT, USA
- Dominion Diagnostics LLC, North Kingstown, Rhode Island, USA
| | - Jan Baumbach
- Computational Biology Group Department of Mathematics and Computer Science, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
39
|
Spinner JL, Jarrett CO, LaRock DL, Miller SI, Collins CM, Hinnebusch BJ. Yersinia pestis insecticidal-like toxin complex (Tc) family proteins: characterization of expression, subcellular localization, and potential role in infection of the flea vector. BMC Microbiol 2012; 12:296. [PMID: 23249165 PMCID: PMC3543167 DOI: 10.1186/1471-2180-12-296] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/12/2012] [Indexed: 12/30/2022] Open
Abstract
Background Toxin complex (Tc) family proteins were first identified as insecticidal toxins in Photorhabdus luminescens and have since been found in a wide range of bacteria. The genome of Yersinia pestis, the causative agent of bubonic plague, contains a locus that encodes the Tc protein homologues YitA, YitB, YitC, and YipA and YipB. Previous microarray data indicate that the Tc genes are highly upregulated by Y. pestis while in the flea vector; however, their role in the infection of fleas and pathogenesis in the mammalian host is unclear. Results We show that the Tc proteins YitA and YipA are highly produced by Y. pestis while in the flea but not during growth in brain heart infusion (BHI) broth at the same temperature. Over-production of the LysR-type regulator YitR from an exogenous plasmid increased YitA and YipA synthesis in broth culture. The increase in production of YitA and YipA correlated with the yitR copy number and was temperature-dependent. Although highly synthesized in fleas, deletion of the Tc proteins did not alter survival of Y. pestis in the flea or prevent blockage of the proventriculus. Furthermore, YipA was found to undergo post-translational processing and YipA and YitA are localized to the outer membrane of Y. pestis. YitA was also detected by immunofluorescence microscopy on the surface of Y. pestis. Both YitA and YipA are produced maximally at low temperature but persist for several hours after transfer to 37°C. Conclusions Y. pestis Tc proteins are highly expressed in the flea but are not essential for Y. pestis to stably infect or produce a transmissible infection in the flea. However, YitA and YipA localize to the outer membrane and YitA is exposed on the surface, indicating that at least YitA is present on the surface when Y. pestis is transmitted into the mammalian host from the flea.
Collapse
Affiliation(s)
- Justin L Spinner
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT 59840, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Yang R, Du Z, Han Y, Zhou L, Song Y, Zhou D, Cui Y. Omics strategies for revealing Yersinia pestis virulence. Front Cell Infect Microbiol 2012; 2:157. [PMID: 23248778 PMCID: PMC3521224 DOI: 10.3389/fcimb.2012.00157] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/27/2012] [Indexed: 01/12/2023] Open
Abstract
Omics has remarkably changed the way we investigate and understand life. Omics differs from traditional hypothesis-driven research because it is a discovery-driven approach. Mass datasets produced from omics-based studies require experts from different fields to reveal the salient features behind these data. In this review, we summarize omics-driven studies to reveal the virulence features of Yersinia pestis through genomics, trascriptomics, proteomics, interactomics, etc. These studies serve as foundations for further hypothesis-driven research and help us gain insight into Y. pestis pathogenesis.
Collapse
Affiliation(s)
- Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology Beijing, China.
| | | | | | | | | | | | | |
Collapse
|
41
|
Uittenbogaard AM, Chelvarajan RL, Myers-Morales T, Gorman AA, Brickey WJ, Ye Z, Kaplan AM, Cohen DA, Ting JPY, Straley SC. Toward a molecular pathogenic pathway for Yersinia pestis YopM. Front Cell Infect Microbiol 2012; 2:155. [PMID: 23248776 PMCID: PMC3518861 DOI: 10.3389/fcimb.2012.00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/22/2012] [Indexed: 11/13/2022] Open
Abstract
YopM is one of the six "effector Yops" of the human-pathogenic Yersinia, but its mechanism has not been defined. After delivery to J774A.1 monocyte-like cells, YopM can rapidly bind and activate the serine/threonine kinases RSK1 and PRK2. However, in infected mice, effects of Y. pestis YopM have been seen only after 24-48 h post-infection (p.i.). To identify potential direct effects of YopM in-vivo we tested for effects of YopM at 1 h and 16-18 h p.i. in mice infected systemically with 10(6) bacteria. At 16 h p.i., there was a robust host response to both parent and ΔyopM-1 Y. pestis KIM5. Compared to cells from non-infected mice, CD11b(+) cells from spleens of infected mice produced more than 100-fold greater IFNγ. In the corresponding sera there were more than 100-fold greater amounts of IFNγ, G-CSF, and CXCL9, as well as more than 10-fold greater amounts of IL-6, CXCL10, and CXCL1. The only YopM-related differences were slightly lower CXCL10 and IL-6 in sera from mice infected 16 h with parent compared to ΔyopM-1 Y. pestis. Microarray analysis of the CD11b(+) cells did not identify consistent transcriptional differences of ≥4-fold at 18 h p.i. However, at 1 h p.i. mRNA for early growth response transcription factor 1 (Egr1) was decreased when YopM was present. Bone marrow-derived macrophages infected for 1 h also expressed lower Egr1 message when YopM was present. Infected J774A.1 cells showed greater expression of Egr1 at 1 h p.i. when YopM was present, but this pattern reversed at 3 h. At 6 h p.i., Cxcl10 mRNA was lower in parent-strain infected cells. We conclude that decreased Egr1 expression is a very early transcriptional effect of YopM and speculate that a pathway may exist from RSK1 through Egr1. These studies revealed novel early transcriptional effects of YopM but point to a time after 18 h of infection when critical transitional events lead to later major effects on cytokine gene transcription.
Collapse
Affiliation(s)
- Annette M Uittenbogaard
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky Lexington, KY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lenz JD, Temple BRS, Miller VL. Evolution and virulence contributions of the autotransporter proteins YapJ and YapK of Yersinia pestis CO92 and their homologs in Y. pseudotuberculosis IP32953. Infect Immun 2012; 80:3693-705. [PMID: 22802344 PMCID: PMC3457547 DOI: 10.1128/iai.00529-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/08/2012] [Indexed: 01/08/2023] Open
Abstract
Yersinia pestis, the causative agent of plague, evolved from the gastrointestinal pathogen Yersinia pseudotuberculosis. Both species have numerous type Va autotransporters, most of which appear to be highly conserved. In Y. pestis CO92, the autotransporter genes yapK and yapJ share a high level of sequence identity. By comparing yapK and yapJ to three homologous genes in Y. pseudotuberculosis IP32953 (YPTB0365, YPTB3285, and YPTB3286), we show that yapK is conserved in Y. pseudotuberculosis, while yapJ is unique to Y. pestis. All of these autotransporters exhibit >96% identity in the C terminus of the protein and identities ranging from 58 to 72% in their N termini. By extending this analysis to include homologous sequences from numerous Y. pestis and Y. pseudotuberculosis strains, we determined that these autotransporters cluster into a YapK (YPTB3285) class and a YapJ (YPTB3286) class. The YPTB3286-like gene of most Y. pestis strains appears to be inactivated, perhaps in favor of maintaining yapJ. Since autotransporters are important for virulence in many bacterial pathogens, including Y. pestis, any change in autotransporter content should be considered for its impact on virulence. Using established mouse models of Y. pestis infection, we demonstrated that despite the high level of sequence identity, yapK is distinct from yapJ in its contribution to disseminated Y. pestis infection. In addition, a mutant lacking both of these genes exhibits an additive attenuation, suggesting nonredundant roles for yapJ and yapK in systemic Y. pestis infection. However, the deletion of the homologous genes in Y. pseudotuberculosis does not seem to impact the virulence of this organism in orogastric or systemic infection models.
Collapse
Affiliation(s)
- Jonathan D. Lenz
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- Division of Biology and Biomedical Sciences, Washington University, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University, St. Louis, Missouri, USA
| | - Brenda R. S. Temple
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
- R. L. Juliano Structural Bioinformatics Core Facility, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Virginia L. Miller
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
- Division of Biology and Biomedical Sciences, Washington University, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
43
|
Kinetics of innate immune response to Yersinia pestis after intradermal infection in a mouse model. Infect Immun 2012; 80:4034-45. [PMID: 22966041 DOI: 10.1128/iai.00606-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A hallmark of Yersinia pestis infection is a delayed inflammatory response early in infection. In this study, we use an intradermal model of infection to study early innate immune cell recruitment. Mice were injected intradermally in the ear with wild-type (WT) or attenuated Y. pestis lacking the pYV virulence plasmid (pYV(-)). The inflammatory responses in ear and draining lymph node samples were evaluated by flow cytometry and immunohistochemistry. As measured by flow cytometry, total neutrophil and macrophage recruitment to the ear in WT-infected mice did not differ from phosphate-buffered saline (PBS) controls or mice infected with pYV(-), except for a transient increase in macrophages at 6 h compared to the PBS control. Limited inflammation was apparent even in animals with high bacterial loads (10(5) to 10(6) CFU). In addition, activation of inflammatory cells was significantly reduced in WT-infected mice as measured by CD11b and major histocompatibility complex class II (MHC-II) expression. When mice infected with WT were injected 12 h later at the same intradermal site with purified LPS, Y. pestis did not prevent recruitment of neutrophils. However, significant reduction in neutrophil activation remained compared to that of PBS and pYV(-) controls. Immunohistochemistry revealed qualitative differences in neutrophil recruitment to the skin and draining lymph node, with WT-infected mice producing a diffuse inflammatory response. In contrast, focal sites of neutrophil recruitment were sustained through 48 h postinfection in pYV(-)-infected mice. Thus, an important feature of Y. pestis infection is reduced activation and organization of inflammatory cells that is at least partially dependent on the pYV virulence plasmid.
Collapse
|
44
|
Williamson ED, Oyston PCF. The natural history and incidence of Yersinia pestis and prospects for vaccination. J Med Microbiol 2012; 61:911-918. [PMID: 22442294 DOI: 10.1099/jmm.0.037960-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Plague is an ancient, serious, infectious disease which is still endemic in regions of the modern world and is a potential biothreat agent. This paper discusses the natural history of the bacterium and its evolution into a flea-vectored bacterium able to transmit bubonic plague. It reviews the incidence of plague in the modern world and charts the history of vaccines which have been used to protect against the flea-vectored disease, which erupts as bubonic plague. Current approaches to vaccine development to protect against pneumonic, as well as bubonic, plague are also reviewed. The considerable challenges in achieving a vaccine which is licensed for human use and which will comprehensively protect against this serious human pathogen are assessed.
Collapse
Affiliation(s)
- E D Williamson
- Biomedical Sciences, Dstl Porton Down, Salisbury SP4 0JQ, UK
| | - P C F Oyston
- Biomedical Sciences, Dstl Porton Down, Salisbury SP4 0JQ, UK
| |
Collapse
|
45
|
Role of the Yersinia pestis Ail protein in preventing a protective polymorphonuclear leukocyte response during bubonic plague. Infect Immun 2011; 79:4984-9. [PMID: 21969002 DOI: 10.1128/iai.05307-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability of Yersinia pestis to forestall the mammalian innate immune response is a fundamental aspect of plague pathogenesis. In this study, we examined the effect of Ail, a 17-kDa outer membrane protein that protects Y. pestis against complement-mediated lysis, on bubonic plague pathogenesis in mice and rats. The Y. pestis ail mutant was attenuated for virulence in both rodent models. The attenuation was greater in rats than in mice, which correlates with the ability of normal rat serum, but not mouse serum, to kill ail-negative Y. pestis in vitro. Intradermal infection with the ail mutant resulted in an atypical, subacute form of bubonic plague associated with extensive recruitment of polymorphonuclear leukocytes (PMN or neutrophils) to the site of infection in the draining lymph node and the formation of large purulent abscesses that contained the bacteria. Systemic spread and mortality were greatly attenuated, however, and a productive adaptive immune response was generated after high-dose challenge, as evidenced by high serum antibody levels against Y. pestis F1 antigen. The Y. pestis Ail protein is an important bubonic plague virulence factor that inhibits the innate immune response, in particular the recruitment of a protective PMN response to the infected lymph node.
Collapse
|
46
|
Resistance to Yersinia pestis infection decreases with age in B10.T(6R) mice. Infect Immun 2011; 79:4438-46. [PMID: 21859850 DOI: 10.1128/iai.05267-11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We demonstrate that 2-month-old female B10.T(6R) mice are highly resistant to systemic infection with the KIM5 strain of Yersinia pestis and that B10.T(6R) mice become susceptible to Y. pestis infection by the age of 5 months. In this study, young (2-month-old) and middle-aged (5- to 12-month-old) B10.T(6R) mice were infected with equal CFU counts of Y. pestis. The 50% lethal dose (LD(50)) for young B10.T(6R) mice was ∼1.4 × 10(4) CFU, while middle-aged B10.T(6R) mice exhibited an LD(50) of ∼60 CFU. Elevated bacterial burdens were found in the spleens of middle-aged mice at 24 and 60 h and in the livers at 60 h postinfection. Immune cell infiltration was greater in the livers of resistant young mice than in those of middle-aged mice and mice of the susceptible C57BL/6N strain. Unlike susceptible mice, young B10.T(6R) mice did not develop necrotic lesions throughout the liver. Instead, livers from young B10.T(6R) mice contained granuloma-like structures. Immunohistochemical staining of liver sections from these mice at 60 h postinfection revealed that the majority of immune cells present in these structures were neutrophils. These findings suggest that resistance to plague in B10.T(6R) mice correlates with early formation of neutrophilic lesions in the liver.
Collapse
|