Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Infect Immun ; 88(1)2019 12 17.
Article in English | MEDLINE | ID: mdl-31636138

ABSTRACT

Salmonella is an intracellular bacterium found in the gastrointestinal tract of mammalian, avian, and reptilian hosts. Mouse models have been extensively used to model in vivo distinct aspects of human Salmonella infections and have led to the identification of several host susceptibility genes. We have investigated the susceptibility of Collaborative Cross strains to intravenous infection with Salmonella enterica serovar Typhimurium as a model of human systemic invasive infection. In this model, strain CC042/GeniUnc (CC042) mice displayed extreme susceptibility with very high bacterial loads and mortality. CC042 mice showed lower spleen weights and decreased splenocyte numbers before and after infection, affecting mostly CD8+ T cells, B cells, and all myeloid cell populations, compared with control C57BL/6J mice. CC042 mice also had lower thymus weights with a reduced total number of thymocytes and double-negative and double-positive (CD4+, CD8+) thymocytes compared to C57BL/6J mice. Analysis of bone marrow-resident hematopoietic progenitors showed a strong bias against lymphoid-primed multipotent progenitors. An F2 cross between CC042 and C57BL/6N mice identified two loci on chromosome 7 (Stsl6 and Stsl7) associated with differences in bacterial loads. In the Stsl7 region, CC042 carried a loss-of-function variant, unique to this strain, in the integrin alpha L (Itgal) gene, the causative role of which was confirmed by a quantitative complementation test. Notably, Itgal loss of function increased the susceptibility to S. Typhimurium in a (C57BL/6J × CC042)F1 mouse background but not in a C57BL/6J mouse inbred background. These results further emphasize the utility of the Collaborative Cross to identify new host genetic variants controlling susceptibility to infections and improve our understanding of the function of the Itgal gene.


Subject(s)
Bacteremia/genetics , CD11a Antigen/deficiency , Genetic Predisposition to Disease , Loss of Function Mutation , Salmonella Infections/genetics , Salmonella typhimurium/growth & development , Animals , Bacteremia/immunology , Bacteremia/pathology , Bacterial Load , Bone Marrow/pathology , Disease Models, Animal , Genes , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/pathology , Serogroup , Spleen/pathology , Survival Analysis , Thymus Gland/pathology
2.
Nat Microbiol ; 4(9): 1516-1531, 2019 09.
Article in English | MEDLINE | ID: mdl-31285585

ABSTRACT

Salmonella presents a global public health concern. Central to Salmonella pathogenicity is an ability to subvert host defences through strategically targeting host proteins implicated in restricting infection. Therefore, to gain insight into the host-pathogen interactions governing Salmonella infection, we performed an in vivo genome-wide mutagenesis screen to uncover key host defence proteins. This revealed an uncharacterized role of CYRI (FAM49B) in conferring host resistance to Salmonella infection. We show that CYRI binds to the small GTPase RAC1 through a conserved domain present in CYFIP proteins, which are known RAC1 effectors that stimulate actin polymerization. However, unlike CYFIP proteins, CYRI negatively regulates RAC1 signalling, thereby attenuating processes such as macropinocytosis, phagocytosis and cell migration. This enables CYRI to counteract Salmonella at various stages of infection, including bacterial entry into non-phagocytic and phagocytic cells as well as phagocyte-mediated bacterial dissemination. Intriguingly, to dampen its effects, the bacterial effector SopE, a RAC1 activator, selectively targets CYRI following infection. Together, this outlines an intricate host-pathogen signalling interplay that is crucial for determining bacterial fate. Notably, our study also outlines a role for CYRI in restricting infection mediated by Mycobacterium tuberculosis and Listeria monocytogenes. This provides evidence implicating CYRI cellular functions in host defence beyond Salmonella infection.


Subject(s)
Bacterial Infections/prevention & control , Cytoskeleton/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Bacterial Infections/metabolism , Bacterial Infections/microbiology , Bacterial Load , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytoskeleton/genetics , Disease Resistance/genetics , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Intracellular Signaling Peptides and Proteins/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/physiology , Macrophages/microbiology , Macrophages/pathology , Mice , Mitochondrial Proteins/genetics , Mutation , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/physiology , Phagocytosis , Protein Binding , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology , Survival Analysis
3.
Nat Immunol ; 17(1): 65-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595887

ABSTRACT

Viral respiratory tract infections are the main causative agents of the onset of infection-induced asthma and asthma exacerbations that remain mechanistically unexplained. Here we found that deficiency in signaling via type I interferon receptor led to deregulated activation of group 2 innate lymphoid cells (ILC2 cells) and infection-associated type 2 immunopathology. Type I interferons directly and negatively regulated mouse and human ILC2 cells in a manner dependent on the transcriptional activator ISGF3 that led to altered cytokine production, cell proliferation and increased cell death. In addition, interferon-γ (IFN-γ) and interleukin 27 (IL-27) altered ILC2 function dependent on the transcription factor STAT1. These results demonstrate that type I and type II interferons, together with IL-27, regulate ILC2 cells to restrict type 2 immunopathology.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Lymphocytes/immunology , Respiratory Tract Infections/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/pathology
4.
Genes (Basel) ; 5(4): 887-925, 2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25268389

ABSTRACT

Infectious diseases are responsible for over 25% of deaths globally, but many more individuals are exposed to deadly pathogens. The outcome of infection results from a set of diverse factors including pathogen virulence factors, the environment, and the genetic make-up of the host. The completion of the human reference genome sequence in 2004 along with technological advances have tremendously accelerated and renovated the tools to study the genetic etiology of infectious diseases in humans and its best characterized mammalian model, the mouse. Advancements in mouse genomic resources have accelerated genome-wide functional approaches, such as gene-driven and phenotype-driven mutagenesis, bringing to the fore the use of mouse models that reproduce accurately many aspects of the pathogenesis of human infectious diseases. Treatment with the mutagen N-ethyl-N-nitrosourea (ENU) has become the most popular phenotype-driven approach. Our team and others have employed mouse ENU mutagenesis to identify host genes that directly impact susceptibility to pathogens of global significance. In this review, we first describe the strategies and tools used in mouse genetics to understand immunity to infection with special emphasis on chemical mutagenesis of the mouse germ-line together with current strategies to efficiently identify functional mutations using next generation sequencing. Then, we highlight illustrative examples of genes, proteins, and cellular signatures that have been revealed by ENU screens and have been shown to be involved in susceptibility or resistance to infectious diseases caused by parasites, bacteria, and viruses.

5.
J Immunol ; 192(1): 259-70, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24285835

ABSTRACT

Salmonella enterica is a ubiquitous Gram-negative intracellular bacterium that continues to pose a global challenge to human health. The etiology of Salmonella pathogenesis is complex and controlled by pathogen, environmental, and host genetic factors. In fact, patients immunodeficient in genes in the IL-12, IL-23/IFN-γ pathway are predisposed to invasive nontyphoidal Salmonella infection. Using a forward genomics approach by N-ethyl-N-nitrosourea (ENU) germline mutagenesis in mice, we identified the Ity14 (Immunity to Typhimurium locus 14) pedigree exhibiting increased susceptibility following in vivo Salmonella challenge. A DNA-binding domain mutation (p.G418_E445) in Stat4 (Signal Transducer and Activator of Transcription Factor 4) was the causative mutation. STAT4 signals downstream of IL-12 to mediate transcriptional regulation of inflammatory immune responses. In mutant Ity14 mice, the increased splenic and hepatic bacterial load resulted from an intrinsic defect in innate cell function, IFN-γ-mediated immunity, and disorganized granuloma formation. We further show that NK and NKT cells play an important role in mediating control of Salmonella in Stat4(Ity14/Ity14) mice. Stat4(Ity14/Ity14) mice had increased expression of genes involved in cell-cell interactions and communication, as well as increased CD11b expression on a subset of splenic myeloid dendritic cells, resulting in compromised recruitment of inflammatory cells to the spleen during Salmonella infection. Stat4(Ity14/Ity14) presented upregulated compensatory mechanisms, although inefficient and ultimately Stat4(Ity14/Ity14) mice develop fatal bacteremia. The following study further elucidates the pathophysiological impact of STAT4 during Salmonella infection.


Subject(s)
Gene Expression Regulation , Genetic Predisposition to Disease , Interferon-gamma/immunology , Mutation , STAT4 Transcription Factor/genetics , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Transcription, Genetic , Animals , Bacterial Load , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cation Transport Proteins/genetics , Cluster Analysis , DNA Mutational Analysis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Loci , Immunity, Innate/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Liver/microbiology , Mice , Mutation/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Nitrosourea Compounds/toxicity , Pedigree , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella typhimurium/immunology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Transcriptome
6.
G3 (Bethesda) ; 3(10): 1753-8, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23979929

ABSTRACT

Using the N-ethyl-N-nitrosourea (ENU) mutagenesis screen, we have identified two skeletal morphology mutants, Skm1 and Skm2. Positional cloning and candidate gene sequencing localized the causative point mutations within the genes coding for natriuretic peptide receptor C (NPR-C) and filamin b (FLNB), respectively. Mice that carry a mutation in Npr3 exhibit a skeletal overgrowth phenotype, resulting in an elongated body and kyphosis. Skm2 mice, carrying a mutation in Flnb, present with scoliosis and lordosis. These mutant mice will serve as useful models for the study of vertebral malformations.


Subject(s)
Bone and Bones/abnormalities , Filamins/genetics , Mutation, Missense , Receptors, Atrial Natriuretic Factor/genetics , Spinal Curvatures/genetics , Amino Acid Sequence , Animals , Ethylnitrosourea , Filamins/metabolism , Mice , Molecular Sequence Data , Mutagens , Receptors, Atrial Natriuretic Factor/metabolism
7.
Mamm Genome ; 24(5-6): 218-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23588612

ABSTRACT

The Gram-negative bacteria, Salmonella, cause a broad spectrum of clinical diseases in humans, ranging from asymptomatic carriage to life-threatening sepsis. We have designed an experimental model to study the contribution of genetic factors to the persistence of Salmonella Enteritidis during the late phase of infection in 129S6/SvEvTac and C57BL/6J mice. C57BL/6J mice cleared the bacteria from their reticuloendothelial system within a period of 42 days, whereas the 129S6 mice still presented a high bacterial load. Using this model, we have identified ten Salmonella Enteritidis susceptibility loci (Ses1, Ses1.1, and Ses3-Ses10) associated with bacterial persistence in target organs of 129S6/SvEvTac mice using a two-locus epistasis QTL linkage mapping approach. Significant statistical interactions were detected between Ses1 on chromosome 1 and Ses5 on chromosome 7 and between Ses1 and Ses4 on chromosome X. In this study, we functionally validated the genetic architecture of Salmonella persistence in 129S6 mice using single- (129S6.B6-Ses1.2 that combines Ses1 and Ses1.1 loci, 129S6.B6-Ses4, and 129S6.B6-Ses5) and double-congenic mice (129S6.B6-Ses1.2/Ses4 and 129S6.B6-Ses1.2/Ses5). These experiments demonstrate functional interactions between Ses1.2 and Ses4 or Ses5 that improve Salmonella Enteritidis clearance, validating the critical role played by gene-gene interactions in the contribution to bacterial clearance heritability. Improved bacterial clearance in double-congenic mice could be explained by the impact of Ses4 and Ses5 in combination with Ses1.2 on TH polarization since a TH2 bias (decreased Ifng and increased Il4 mRNA levels and reduced IgG2a immunoglobulins in the serum) was observed in 129S6.B6-Ses1.2/Ses5 mice and a TH17 (high Il17 expression) bias in 129S6.B6-Ses1.2/Ses4.


Subject(s)
Mice/genetics , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella enteritidis/physiology , Animals , Chromosome Mapping , Disease Models, Animal , Female , Humans , Male , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred Strains
8.
PLoS One ; 8(2): e55331, 2013.
Article in English | MEDLINE | ID: mdl-23390527

ABSTRACT

Salmonella, a ubiquitous Gram-negative intracellular bacterium, is a food borne pathogen that infects a broad range of hosts. Infection with Salmonella Typhimurium in mice is a broadly recognized experimental model resembling typhoid fever in humans. Using a N-ethyl-N-nitrosurea (ENU) mutagenesis recessive screen, we report the identification of Ity16 (Immunity to Typhimurium locus 16), a locus responsible for increased susceptibility to infection. The position of Ity16 was refined on chromosome 8 and a nonsense mutation was identified in the ankyrin 1 (Ank1) gene. ANK1 plays an important role in the formation and stabilization of the red cell cytoskeleton. The Ank1(Ity16/Ity16) mutation causes severe hemolytic anemia in uninfected mice resulting in splenomegaly, hyperbilirubinemia, jaundice, extramedullary erythropoiesis and iron overload in liver and kidneys. Ank1(Ity16/Ity16) mutant mice demonstrated low levels of hepcidin (Hamp) expression and significant increases in the expression of the growth differentiation factor 15 (Gdf15), erythropoietin (Epo) and heme oxygenase 1 (Hmox1) exacerbating extramedullary erythropoiesis, tissue iron deposition and splenomegaly. As the infection progresses in Ank1(Ity16/Ity16), the anemia worsens and bacterial load were high in liver and kidneys compared to wild type mice. Heterozygous Ank1(+/Ity16) mice were also more susceptible to Salmonella infection although to a lesser extent than Ank1(Ity16/Ity16) and they did not inherently present anemia and splenomegaly. During infection, iron accumulated in the kidneys of Ank1(+/Ity16) mice where bacterial loads were high compared to littermate controls. The critical role of HAMP in the host response to Salmonella infection was validated by showing increased susceptibility to infection in Hamp-deficient mice and significant survival benefits in Ank1(+/Ity16) heterozygous mice treated with HAMP peptide. This study illustrates that the regulation of Hamp and iron balance are crucial in the host response to Salmonella infection in Ank1 mutants.


Subject(s)
Anemia, Hemolytic/genetics , Ankyrins/genetics , Antimicrobial Cationic Peptides/genetics , Codon, Nonsense/drug effects , Ethylnitrosourea/toxicity , Iron Overload/genetics , Salmonella Infections/genetics , Anemia, Hemolytic/metabolism , Anemia, Hemolytic/microbiology , Anemia, Hemolytic/mortality , Animals , Ankyrins/metabolism , Antimicrobial Cationic Peptides/deficiency , Erythrocytes/metabolism , Erythrocytes/pathology , Erythropoietin/genetics , Erythropoietin/metabolism , Gene Expression , Genetic Predisposition to Disease , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hepcidins , Heterozygote , Homozygote , Iron/metabolism , Iron Overload/metabolism , Iron Overload/microbiology , Iron Overload/mortality , Liver/metabolism , Liver/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/mortality , Salmonella typhimurium/physiology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...