Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Heredity (Edinb) ; 128(4): 250-260, 2022 04.
Article in English | MEDLINE | ID: mdl-35256765

ABSTRACT

The genetic basis of traits shapes and constrains how adaptation proceeds in nature; rapid adaptation can proceed using stores of polygenic standing genetic variation or hard selective sweeps, and increasing polygenicity fuels genetic redundancy, reducing gene re-use (genetic convergence). Guppy life history traits evolve rapidly and convergently among natural high- and low-predation environments in northern Trinidad. This system has been studied extensively at the phenotypic level, but little is known about the underlying genetic architecture. Here, we use four independent F2 QTL crosses to examine the genetic basis of seven (five female, two male) guppy life history phenotypes and discuss how these genetic architectures may facilitate or constrain rapid adaptation and convergence. We use RAD-sequencing data (16,539 SNPs) from 370 male and 267 female F2 individuals. We perform linkage mapping, estimates of genome-wide and per-chromosome heritability (multi-locus associations), and QTL mapping (single-locus associations). Our results are consistent with architectures of many loci of small-effect for male age and size at maturity and female interbrood period. Male trait associations are clustered on specific chromosomes, but female interbrood period exhibits a weak genome-wide signal suggesting a potentially highly polygenic component. Offspring weight and female size at maturity are also associated with a single significant QTL each. These results suggest rapid, repeatable phenotypic evolution of guppies may be facilitated by polygenic trait architectures, but subsequent genetic redundancy may limit gene re-use across populations, in agreement with an absence of strong signatures of genetic convergence from recent analyses of wild guppies.


Subject(s)
Life History Traits , Poecilia , Animals , Chromosome Mapping , Female , Male , Multifactorial Inheritance , Phenotype , Poecilia/genetics , Quantitative Trait Loci
2.
Cell Host Microbe ; 28(1): 54-68.e7, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32526160

ABSTRACT

The Salmonella enterica effector SteD depletes mature MHC class II (mMHCII) molecules from the surface of infected antigen-presenting cells through ubiquitination of the cytoplasmic tail of the mMHCII ß chain. Here, through a genome-wide mutant screen of human antigen-presenting cells, we show that the NEDD4 family HECT E3 ubiquitin ligase WWP2 and a tumor-suppressing transmembrane protein of unknown biochemical function, TMEM127, are required for SteD-dependent ubiquitination of mMHCII. Although evidently not involved in normal regulation of mMHCII, TMEM127 was essential for SteD to suppress both mMHCII antigen presentation in mouse dendritic cells and MHCII-dependent CD4+ T cell activation. We found that TMEM127 contains a canonical PPxY motif, which was required for binding to WWP2. SteD bound to TMEM127 and enabled TMEM127 to interact with and induce ubiquitination of mature MHCII. Furthermore, SteD also underwent TMEM127- and WWP2-dependent ubiquitination, which both contributed to its degradation and augmented its activity on mMHCII.


Subject(s)
Bacterial Proteins/physiology , Histocompatibility Antigens Class II/metabolism , Membrane Proteins/physiology , Salmonella typhimurium/physiology , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Animals , Antigen Presentation , CRISPR-Cas Systems , Cell Line , Dendritic Cells/immunology , Dendritic Cells/microbiology , Female , Host-Pathogen Interactions , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mutation , Protein Binding , Salmonella Infections/immunology , Salmonella Infections/microbiology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/microbiology , Virulence
3.
Int J Health Plann Manage ; 34(1): e844-e859, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30402892

ABSTRACT

Sustainable Development Goal 3 (SDG 3) focuses on health and well-being. To understand the in-country monitoring challenges for developing countries of reporting against SDG 3, this research sought published data for the four Pacific countries of Fiji, Papua New Guinea (PNG), the Solomon Islands, and Vanuatu-within a region with well-documented and significant health challenges. This research found that there are limited recent, comprehensive, and comparable data with identified sources against the SDG 3 outcome indicators at an in-country level. Without such data, there is a risk of relying on data that may be inaccurate because of aggregation, estimation, and modelling. The results from these data can influence the funding and other resources that could be made available to the Melanesian countries to address health inequities.


Subject(s)
Health Planning/organization & administration , Population Health , Sustainable Development , Databases, Factual , Goals , Health Status , Humans , Melanesia , United Nations
5.
Infect Immun ; 85(3)2017 03.
Article in English | MEDLINE | ID: mdl-28069818

ABSTRACT

Within host cells such as macrophages, Salmonella enterica translocates virulence (effector) proteins across its vacuolar membrane via the SPI-2 type III secretion system. Previously, it was shown that when expressed ectopically, the effectors SseK1 and SseK3 inhibit tumor necrosis factor alpha (TNF-α)-induced NF-κB activation. In this study, we show that ectopically expressed SseK1, SseK2, and SseK3 suppress TNF-α-induced, but not Toll-like receptor 4- or interleukin-induced, NF-κB activation. Inhibition required a DXD motif in SseK1 and SseK3, which is essential for the transfer of N-acetylglucosamine to arginine residues (arginine-GlcNAcylation). During macrophage infection, SseK1 and SseK3 inhibited NF-κB activity in an additive manner. SseK3-mediated inhibition of NF-κB activation did not require the only known host-binding partner of this effector, the E3-ubiquitin ligase TRIM32. SseK proteins also inhibited TNF-α-induced cell death during macrophage infection. Despite SseK1 and SseK3 inhibiting TNF-α-induced apoptosis upon ectopic expression in HeLa cells, the percentage of infected macrophages undergoing apoptosis was SseK independent. Instead, SseK proteins inhibited necroptotic cell death during macrophage infection. SseK1 and SseK3 caused GlcNAcylation of different proteins in infected macrophages, suggesting that these effectors have distinct substrate specificities. Indeed, SseK1 caused the GlcNAcylation of the death domain-containing proteins FADD and TRADD, whereas SseK3 expression resulted in weak GlcNAcylation of TRADD but not FADD. Additional, as-yet-unidentified substrates are likely to explain the additive phenotype of a Salmonella strain lacking both SseK1 and SseK3.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , NF-kappa B/metabolism , Salmonella/physiology , Signal Transduction , Type III Secretion Systems , Animals , Apoptosis , Arginine/metabolism , Bacterial Proteins/genetics , Cell Death , Cell Line , Cells, Cultured , Gene Knockout Techniques , Glycosylation , HeLa Cells , Host-Pathogen Interactions , Humans , Mice , Protein Binding , Protein Transport , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
Nat Commun ; 7: 13292, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27808091

ABSTRACT

Sensing bacterial products in the cytosol of mammalian cells by NOD-like receptors leads to the activation of caspase-1 inflammasomes, and the production of the pro-inflammatory cytokines interleukin (IL)-18 and IL-1ß. In addition, mouse caspase-11 (represented in humans by its orthologs, caspase-4 and caspase-5) detects cytosolic bacterial LPS directly. Activation of caspase-1 and caspase-11 initiates pyroptotic host cell death that releases potentially harmful bacteria from the nutrient-rich host cell cytosol into the extracellular environment. Here we use single cell analysis and time-lapse microscopy to identify a subpopulation of host cells, in which growth of cytosolic Salmonella Typhimurium is inhibited independently or prior to the onset of cell death. The enzymatic activities of caspase-1 and caspase-11 are required for growth inhibition in different cell types. Our results reveal that these proteases have important functions beyond the direct induction of pyroptosis and proinflammatory cytokine secretion in the control of growth and elimination of cytosolic bacteria.


Subject(s)
Caspase 1/immunology , Caspases/immunology , Cytosol/immunology , Pyroptosis/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , 3T3 Cells , Animals , Caspase 1/genetics , Caspase 1/metabolism , Caspases/genetics , Caspases/metabolism , Caspases, Initiator , Cytosol/enzymology , Cytosol/microbiology , Disease Models, Animal , Extracellular Space/microbiology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Inflammasomes/immunology , Inflammasomes/metabolism , Macrophages/enzymology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/microbiology , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , Single-Cell Analysis , Time-Lapse Imaging
7.
EMBO J ; 35(16): 1779-92, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27370208

ABSTRACT

Mammalian cells deploy autophagy to defend their cytosol against bacterial invaders. Anti-bacterial autophagy relies on the core autophagy machinery, cargo receptors, and "eat-me" signals such as galectin-8 and ubiquitin that label bacteria as autophagy cargo. Anti-bacterial autophagy also requires the kinase TBK1, whose role in autophagy has remained enigmatic. Here we show that recruitment of WIPI2, itself essential for anti-bacterial autophagy, is dependent on the localization of catalytically active TBK1 to the vicinity of cytosolic bacteria. Experimental manipulation of TBK1 recruitment revealed that engagement of TBK1 with any of a variety of Salmonella-associated "eat-me" signals, including host-derived glycans and K48- and K63-linked ubiquitin chains, suffices to restrict bacterial proliferation. Promiscuity in recruiting TBK1 via independent signals may buffer TBK1 functionality from potential bacterial antagonism and thus be of evolutionary advantage to the host.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Cytosol/microbiology , Immunity, Innate , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Salmonella typhimurium/immunology , Animals , Humans , Mice , Phosphate-Binding Proteins
8.
PLoS Pathog ; 12(5): e1005653, 2016 05.
Article in English | MEDLINE | ID: mdl-27232334

ABSTRACT

Salmonella enterica replicates in macrophages through the action of effector proteins translocated across the vacuolar membrane by a type III secretion system (T3SS). Here we show that the SPI-2 T3SS effector SpvD suppresses proinflammatory immune responses. SpvD prevented activation of an NF-ĸB-dependent promoter and caused nuclear accumulation of importin-α, which is required for nuclear import of p65. SpvD interacted specifically with the exportin Xpo2, which mediates nuclear-cytoplasmic recycling of importins. We propose that interaction between SpvD and Xpo2 disrupts the normal recycling of importin-α from the nucleus, leading to a defect in nuclear translocation of p65 and inhibition of activation of NF-ĸB regulated promoters. SpvD down-regulated pro-inflammatory responses and contributed to systemic growth of bacteria in mice. This work shows that a bacterial pathogen can manipulate host cell immune responses by interfering with the nuclear transport machinery.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions/physiology , Salmonella Infections, Animal/metabolism , Transcription Factor RelA/metabolism , Virulence Factors/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RAW 264.7 Cells , Salmonella Infections, Animal/immunology , Salmonella enterica/immunology , Type III Secretion Systems/metabolism , Virulence Factors/immunology
9.
Science ; 343(6167): 204-8, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24408438

ABSTRACT

Many bacterial pathogens cause persistent infections despite repeated antibiotic exposure. Bacterial persisters are antibiotic-tolerant cells, but little is known about their growth status and the signals and pathways leading to their formation in infected tissues. We used fluorescent single-cell analysis to identify Salmonella persisters during infection. These were part of a nonreplicating population formed immediately after uptake by macrophages and were induced by vacuolar acidification and nutritional deprivation, conditions that also induce Salmonella virulence gene expression. The majority of 14 toxin-antitoxin modules contributed to intracellular persister formation. Some persisters resumed intracellular growth after phagocytosis by naïve macrophages. Thus, the vacuolar environment induces phenotypic heterogeneity, leading to either bacterial replication or the formation of nonreplicating persisters that could provide a reservoir for relapsing infection.


Subject(s)
Macrophages/microbiology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/growth & development , Animals , Anti-Bacterial Agents/pharmacology , Antitoxins/genetics , Bacterial Toxins/genetics , Cefotaxime/pharmacology , Gene Deletion , Gene Expression Regulation, Bacterial , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mesentery/immunology , Mesentery/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Operon/genetics , Phagocytosis , Pyrophosphatases/genetics , Recurrence , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Spleen/immunology , Spleen/microbiology , Virulence
10.
PLoS One ; 8(12): e84512, 2013.
Article in English | MEDLINE | ID: mdl-24376817

ABSTRACT

The multi-protein ß-barrel assembly machine (BAM) of Escherichia coli is responsible for the folding and insertion of ß-barrel containing integral outer membrane proteins (OMPs) into the bacterial outer membrane. An essential component of this complex is the BamA protein, which binds unfolded ß-barrel precursors via the five polypeptide transport-associated (POTRA) domains in its N-terminus. The C-terminus of BamA contains a ß-barrel domain, which tethers BamA to the outer membrane and is also thought to be involved in OMP insertion. Here we mutagenize BamA using linker scanning mutagenesis and demonstrate that all five POTRA domains are essential for BamA protein function in our experimental system. Furthermore, we generate a homology based model of the BamA ß-barrel and test our model using insertion mutagenesis, deletion analysis and immunofluorescence to identify ß-strands, periplasmic turns and extracellular loops. We show that the surface-exposed loops of the BamA ß-barrel are essential.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Models, Molecular , Bacterial Outer Membrane Proteins/metabolism , Blotting, Western , Escherichia coli Proteins/metabolism , Fluorescent Antibody Technique , Mutagenesis , Plasmids/genetics , Protein Structure, Tertiary
11.
Science ; 338(6109): 963-7, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23162002

ABSTRACT

Salmonella enterica is an intracellular bacterial pathogen that replicates within membrane-bound vacuoles through the action of effector proteins translocated into host cells. Salmonella vacuoles have characteristics of lysosomes but are reduced in hydrolytic enzymes transported by mannose-6-phosphate receptors (MPRs). We found that the effector SifA subverted Rab9-dependent retrograde trafficking of MPRs, thereby attenuating lysosome function. This required binding of SifA to its host cell target SKIP/PLEKHM2. Furthermore, SKIP regulated retrograde trafficking of MPRs in noninfected cells. Translocated SifA formed a stable complex with SKIP and Rab9 in infected cells. Sequestration of Rab9 by SifA-SKIP accounted for the effect of SifA on MPR transport and lysosome function. Growth of Salmonella increased in cells with reduced lysosomal activity and decreased in cells with higher lysosomal activity. These results suggest that Salmonella vacuoles undergo fusion with lysosomes whose potency has been reduced by SifA.


Subject(s)
Bacterial Proteins/metabolism , Glycoproteins/metabolism , Lysosomes/metabolism , Receptor, IGF Type 2/metabolism , Salmonella enterica/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , HeLa Cells , Humans , Membrane Fusion , Protein Transport , RNA, Small Interfering/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 103(40): 14941-6, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-16990433

ABSTRACT

Several pathogenic strains of Escherichia coli exploit type III secretion to inject "effector proteins" into human cells, which then subvert eukaryotic cell biology to the bacterium's advantage. We have exploited bioinformatics and experimental approaches to establish that the effector repertoire in the Sakai strain of enterohemorrhagic E. coli (EHEC) O157:H7 is much larger than previously thought. Homology searches led to the identification of >60 putative effector genes. Thirteen of these were judged to be likely pseudogenes, whereas 49 were judged to be potentially functional. In total, 39 proteins were confirmed experimentally as effectors: 31 through proteomics and 28 through translocation assays. At the protein level, the EHEC effector sequences fall into >20 families. The largest family, the NleG family, contains 14 members in the Sakai strain alone. EHEC also harbors functional homologs of effectors from plant pathogens (HopPtoH, HopW, AvrA) and from Shigella (OspD, OspE, OspG), and two additional members of the Map/IpgB family. Genes encoding proven or predicted effectors occur in >20 exchangeable effector loci scattered throughout the chromosome. Crucially, the majority of functional effector genes are encoded by nine exchangeable effector loci that lie within lambdoid prophages. Thus, type III secretion in E. coli is linked to a vast phage "metagenome," acting as a crucible for the evolution of pathogenicity.


Subject(s)
Bacteriophage lambda/metabolism , Escherichia coli O157/metabolism , Escherichia coli Proteins/metabolism , Chromosomes, Bacterial/genetics , Genome, Bacterial/genetics , Humans , Prophages/genetics , Sequence Homology , Shigella/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...