Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 184(12): 3178-3191.e18, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34022140

ABSTRACT

Gasdermin B (GSDMB) belongs to a large family of pore-forming cytolysins that execute inflammatory cell death programs. While genetic studies have linked GSDMB polymorphisms to human disease, its function in the immunological response to pathogens remains poorly understood. Here, we report a dynamic host-pathogen conflict between GSDMB and the IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri. We show that IpaH7.8 ubiquitinates and targets GSDMB for 26S proteasome destruction. This virulence strategy protects Shigella from the bacteriocidic activity of natural killer cells by suppressing granzyme-A-mediated activation of GSDMB. In contrast to the canonical function of most gasdermin family members, GSDMB does not inhibit Shigella by lysing host cells. Rather, it exhibits direct microbiocidal activity through recognition of phospholipids found on Gram-negative bacterial membranes. These findings place GSDMB as a central executioner of intracellular bacterial killing and reveal a mechanism employed by pathogens to counteract this host defense system.


Subject(s)
Biomarkers, Tumor/metabolism , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Neoplasm Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Shigella flexneri/physiology , Ubiquitination , Animals , Bacterial Proteins/metabolism , Cardiolipins/metabolism , Cell Line , Cell Membrane/metabolism , Female , Granzymes/metabolism , Humans , Lipid A/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Substrate Specificity
2.
PLoS Pathog ; 20(5): e1012010, 2024 May.
Article in English | MEDLINE | ID: mdl-38753575

ABSTRACT

Arboviruses are a diverse group of insect-transmitted pathogens that pose global public health challenges. Identifying evolutionarily conserved host factors that combat arbovirus replication in disparate eukaryotic hosts is important as they may tip the balance between productive and abortive viral replication, and thus determine virus host range. Here, we exploit naturally abortive arbovirus infections that we identified in lepidopteran cells and use bacterial effector proteins to uncover host factors restricting arbovirus replication. Bacterial effectors are proteins secreted by pathogenic bacteria into eukaryotic hosts cells that can inhibit antimicrobial defenses. Since bacteria and viruses can encounter common host defenses, we hypothesized that some bacterial effectors may inhibit host factors that restrict arbovirus replication in lepidopteran cells. Thus, we used bacterial effectors as molecular tools to identify host factors that restrict four distinct arboviruses in lepidopteran cells. By screening 210 effectors encoded by seven different bacterial pathogens, we identify several effectors that individually rescue the replication of all four arboviruses. We show that these effectors encode diverse enzymatic activities that are required to break arbovirus restriction. We further characterize Shigella flexneri-encoded IpaH4 as an E3 ubiquitin ligase that directly ubiquitinates two evolutionarily conserved proteins, SHOC2 and PSMC1, promoting their degradation in insect and human cells. We show that depletion of either SHOC2 or PSMC1 in insect or human cells promotes arbovirus replication, indicating that these are ancient virus restriction factors conserved across invertebrate and vertebrate hosts. Collectively, our study reveals a novel pathogen-guided approach to identify conserved antimicrobial machinery, new effector functions, and conserved roles for SHOC2 and PSMC1 in virus restriction.


Subject(s)
Bacterial Proteins , Host-Pathogen Interactions , Virus Replication , Animals , Virus Replication/physiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Arboviruses , Shigella flexneri/pathogenicity , Arbovirus Infections/virology , Cell Line
3.
Nature ; 532(7599): 394-7, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27007849

ABSTRACT

Endoplasmic reticulum (ER) stress is a major contributor to inflammatory diseases, such as Crohn disease and type 2 diabetes. ER stress induces the unfolded protein response, which involves activation of three transmembrane receptors, ATF6, PERK and IRE1α. Once activated, IRE1α recruits TRAF2 to the ER membrane to initiate inflammatory responses via the NF-κB pathway. Inflammation is commonly triggered when pattern recognition receptors (PRRs), such as Toll-like receptors or nucleotide-binding oligomerization domain (NOD)-like receptors, detect tissue damage or microbial infection. However, it is not clear which PRRs have a major role in inducing inflammation during ER stress. Here we show that NOD1 and NOD2, two members of the NOD-like receptor family of PRRs, are important mediators of ER-stress-induced inflammation in mouse and human cells. The ER stress inducers thapsigargin and dithiothreitol trigger production of the pro-inflammatory cytokine IL-6 in a NOD1/2-dependent fashion. Inflammation and IL-6 production triggered by infection with Brucella abortus, which induces ER stress by injecting the type IV secretion system effector protein VceC into host cells, is TRAF2, NOD1/2 and RIP2-dependent and can be reduced by treatment with the ER stress inhibitor tauroursodeoxycholate or an IRE1α kinase inhibitor. The association of NOD1 and NOD2 with pro-inflammatory responses induced by the IRE1α/TRAF2 signalling pathway provides a novel link between innate immunity and ER-stress-induced inflammation.


Subject(s)
Endoplasmic Reticulum Stress , Inflammation/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Animals , Bacterial Outer Membrane Proteins/metabolism , Brucella abortus/immunology , Brucella abortus/pathogenicity , Cell Line , Dithiothreitol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Female , Humans , Immunity, Innate , Inflammation/chemically induced , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/metabolism , Taurochenodeoxycholic Acid/pharmacology , Thapsigargin/pharmacology , Unfolded Protein Response/drug effects
4.
Infect Immun ; 86(4)2018 04.
Article in English | MEDLINE | ID: mdl-29339461

ABSTRACT

The enteric attaching and effacing (A/E) pathogens enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC) and the invasive pathogens enteroinvasive E. coli (EIEC) and Shigella encode type III secretion systems (T3SS) used to inject effector proteins into human host cells during infection. Among these are a group of effectors required for NF-κB-mediated host immune evasion. Recent studies have identified several effector proteins from A/E pathogens and EIEC/Shigella that are involved in suppression of NF-κB and have uncovered their cellular and molecular functions. A novel mechanism among these effectors from both groups of pathogens is to coordinate effector function during infection. This cooperativity among effector proteins explains how bacterial pathogens are able to effectively suppress innate immune defense mechanisms in response to diverse classes of immune receptor signaling complexes (RSCs) stimulated during infection.


Subject(s)
Bacterial Proteins/immunology , Escherichia coli Proteins/immunology , Escherichia coli/physiology , Host-Pathogen Interactions/immunology , Immunomodulation , Shigella/physiology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Nucleus/metabolism , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , NF-kappa B/metabolism , Protein Transport , Signal Transduction , Type III Secretion Systems , Ubiquitin/metabolism
5.
EMBO J ; 33(5): 403-4, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24480478

ABSTRACT

OspG is a secreted effector kinase from the human pathogen Shigella that is required for the reduction of immune responses during Shigella infection. A new study in The EMBO Journal provides a co-crystal structure of OspG bound to UbcH5c~Ub, revealing how a bacterial kinase can be activated by the host ubiquitin conjugation machinery. These results provide molecular insight into an enigmatic microbial virulence factor that thwarts the host immune surveillance system to cause disease.


Subject(s)
Protein Kinases/metabolism , Shigella flexneri/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Virulence Factors/metabolism , Animals , Humans
6.
Annu Rev Microbiol ; 65: 523-41, 2011.
Article in English | MEDLINE | ID: mdl-21939378

ABSTRACT

Brucellosis is a zoonotic infection caused primarily by the bacterial pathogens Brucella melitensis and B. abortus. It is acquired by consumption of unpasteurized dairy products or by contact with infected animals. Globally, it is one of the most widespread zoonoses, with 500,000 new cases reported each year. In endemic areas, Brucella infections represent a serious public health problem that results in significant morbidity and economic losses. An important feature of the disease is persistent bacterial colonization of the reticuloendothelial system. In this review we discuss recent insights into mechanisms of intracellular survival and immune evasion that contribute to systemic persistence by the pathogenic Brucella species.


Subject(s)
Brucella/physiology , Brucellosis/microbiology , Host-Pathogen Interactions , Zoonoses/microbiology , Animals , Brucella/genetics , Brucella/immunology , Brucella/isolation & purification , Brucellosis/epidemiology , Brucellosis/immunology , Brucellosis/transmission , Humans , Immune Evasion , Public Health , Zoonoses/epidemiology , Zoonoses/transmission
7.
bioRxiv ; 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38352400

ABSTRACT

Arboviruses are a diverse group of insect-transmitted pathogens that pose global public health challenges. Identifying evolutionarily conserved host factors that combat arbovirus replication in disparate eukaryotic hosts is important as they may tip the balance between productive and abortive viral replication, and thus determine virus host range. Here, we exploit naturally abortive arbovirus infections that we identified in lepidopteran cells and use bacterial effector proteins to uncover host factors restricting arbovirus replication. Bacterial effectors are proteins secreted by pathogenic bacteria into eukaryotic hosts cells that can inhibit antimicrobial defenses. Since bacteria and viruses can encounter common host defenses, we hypothesized that some bacterial effectors may inhibit host factors that restrict arbovirus replication in lepidopteran cells. Thus, we used bacterial effectors as molecular tools to identify host factors that restrict four distinct arboviruses in lepidopteran cells. By screening 210 effectors encoded by seven different bacterial pathogens, we identify six effectors that individually rescue the replication of all four arboviruses. We show that these effectors encode diverse enzymatic activities that are required to break arbovirus restriction. We further characterize Shigella flexneri-encoded IpaH4 as an E3 ubiquitin ligase that directly ubiquitinates two evolutionarily conserved proteins, SHOC2 and PSMC1, promoting their degradation in insect and human cells. We show that depletion of either SHOC2 or PSMC1 in insect or human cells promotes arbovirus replication, indicating that these are ancient virus restriction factors conserved across invertebrate and vertebrate hosts. Collectively, our study reveals a novel pathogen-guided approach to identify conserved antimicrobial machinery, new effector functions, and conserved roles for SHOC2 and PSMC1 in virus restriction.

8.
Cell Microbiol ; 12(9): 1195-202, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20670294

ABSTRACT

In humans, pathogenic Brucella species cause a febrile illness known as brucellosis. A key pathogenic trait of this group of organisms is their ability to survive in immune cells and persist in tissues of the reticuloendothelial system, a process that requires the function of a Type IV secretion system. In contrast to other well-studied Gram-negative bacteria, Brucella spp. do not cause inflammation at the site of invasion, but have a latency period of 2-4 weeks before the onset of symptoms. This review discusses several mechanisms that allow Brucella spp. both to evade detection by pattern recognition receptors of the innate immune system and suppress their signalling. In contrast to these stealth features, the VirB Type IV secretion system, which mediates survival within phagocytic cells, stimulates innate immune responses in vivo. The responses stimulated by this virulence factor are sufficient to check bacterial growth, but not to elicit sterilizing immunity. The result is a stand-off between host and pathogen that results in persistent infection.


Subject(s)
Bacterial Secretion Systems , Brucella/immunology , Brucellosis/immunology , Brucellosis/microbiology , Immune Evasion , Virulence Factors/metabolism , Animals , Brucella/pathogenicity , Brucellosis/veterinary , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Virulence
9.
Mol Microbiol ; 70(6): 1378-96, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19019140

ABSTRACT

Survival and replication inside host cells by Brucella spp. requires a type IV secretion system (T4SS), encoded by the virB locus. However, the identity of the molecules secreted by the T4SS has remained elusive. We hypothesized that proteins translocated by the T4SS would be co-regulated with the virB operon. The LuxR family regulator VjbR, known to regulate virB, bound a fragment of the virB promoter containing an 18 bp palindromic motif (virB promoter box), showing that VjbR regulated the virB operon directly. To identify virB co-regulated genes, we searched the Brucella suis 1330 and B. abortus 2308 genomes for genes with an upstream virB promoter box. One hundred and forty-four promoters in the two genomes contained the virB promoter box, including those of fliC encoding flagellin and cgs encoding cyclic beta-glucan synthetase. Thirteen of these proteins were tested for VirB-dependent translocation into macrophages using a beta-lactamase reporter assay. This analysis resulted in the identification of the proteins encoded by BAB1_1652 (VceA) and BR1038/BAB1_1058 (VceC) as novel protein substrates of the Brucella T4SS. VceC could also be translocated by the Legionella pneumophila Dot/Icm T4SS into host cells. Our results suggest that VjbR co-ordinates expression of the T4SS and at least two of its secreted substrates.


Subject(s)
Bacterial Proteins/metabolism , Brucella abortus/metabolism , Brucella suis/metabolism , Macrophages/metabolism , Regulon , Secretory Pathway , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Base Sequence , Brucella abortus/genetics , Brucella suis/genetics , Cell Line , Consensus Sequence , Electrophoretic Mobility Shift Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Macrophages/microbiology , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Protein Transport , Secretory Pathway/genetics
10.
mBio ; 10(4)2019 07 23.
Article in English | MEDLINE | ID: mdl-31337727

ABSTRACT

Subversion of endoplasmic reticulum (ER) function is a feature shared by multiple intracellular bacteria and viruses, and in many cases this disruption of cellular function activates pathways of the unfolded protein response (UPR). In the case of infection with Brucella abortus, the etiologic agent of brucellosis, the unfolded protein response in the infected placenta contributes to placentitis and abortion, leading to pathogen transmission. Here we show that B. abortus infection of pregnant mice led to death of infected placental trophoblasts in a manner that depended on the VirB type IV secretion system (T4SS) and its effector VceC. The trophoblast death program required the ER stress-induced transcription factor CHOP. While NOD1/NOD2 expression in macrophages contributed to ER stress-induced inflammation, these receptors did not play a role in trophoblast death. Both placentitis and abortion were independent of apoptosis-associated Speck-like protein containing a caspase activation and recruitment domain (ASC). These studies show that B. abortus uses its T4SS to induce cell-type-specific responses to ER stress in trophoblasts that trigger placental inflammation and abortion. Our results suggest further that in B. abortus the T4SS and its effectors are under selection as bacterial transmission factors.IMPORTANCEBrucella abortus infects the placenta of pregnant cows, where it replicates to high levels and triggers abortion of the calf. The aborted material is highly infectious and transmits infection to both cows and humans, but very little is known about how B. abortus causes abortion. By studying this infection in pregnant mice, we discovered that B. abortus kills trophoblasts, which are important cells for maintaining pregnancy. This killing required an injected bacterial protein (VceC) that triggered an endoplasmic reticulum (ER) stress response in the trophoblast. By inhibiting ER stress or infecting mice that lack CHOP, a protein induced by ER stress, we could prevent death of trophoblasts, reduce inflammation, and increase the viability of the pups. Our results suggest that B. abortus injects VceC into placental trophoblasts to promote its transmission by abortion.


Subject(s)
Brucella abortus/pathogenicity , Cell Death , Endoplasmic Reticulum Stress , Placenta/microbiology , Trophoblasts/microbiology , Type IV Secretion Systems/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Placenta/cytology , Pregnancy , Transcription Factor CHOP/genetics , Trophoblasts/pathology , Unfolded Protein Response
11.
J Bacteriol ; 190(13): 4427-36, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469100

ABSTRACT

The Brucella abortus virB locus contains 12 open reading frames, termed virB1 through virB12, which encode a type IV secretion system. Polar mutations in the virB locus markedly reduce the ability of B. abortus to survive in cultured macrophages or to persist in organs of mice. While a nonpolar deletion of the virB2 gene reduces survival in cultured macrophages and in organs of mice, a nonpolar deletion of virB1 only reduces survival in macrophages, whereas virB12 is dispensable for either virulence trait. Here we investigated the role of the remaining genes in the virB locus during survival in macrophages and virulence in mice. Mutants carrying nonpolar deletions of the virB3, virB4, virB5, virB6, virB7, virB8, virB9, virB10, or virB11 gene were constructed and characterized. All mutations reduced the ability of B. abortus to survive in J774A.1 mouse macrophage-like cells to a degree similar to that caused by a deletion of the entire virB locus. Deletion of virB3, virB4, virB5, virB6, virB8, virB9, virB10, or virB11 markedly reduced the ability of B. abortus to persist in the spleens of mice at 8 weeks after infection. Interestingly, deletion of virB7 did not reduce the ability of B. abortus to persist in spleens of mice. We conclude that virB2, virB3, virB4, virB5, virB6, virB8, virB9, virB10, and virB11 are essential for virulence of B. abortus in mice, while functions encoded by the virB1, virB7, and virB12 genes are not required for persistence in organs with this animal model.


Subject(s)
Bacterial Proteins/genetics , Brucella/genetics , Gene Expression Regulation, Bacterial , Mononuclear Phagocyte System/microbiology , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Blotting, Western , Brucella/pathogenicity , Brucella abortus/genetics , Brucella abortus/pathogenicity , Cell Line , Female , Genes, Bacterial , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mutation , Operon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/microbiology , Virulence/genetics
12.
Nat Microbiol ; 1(7): 16084, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27572974

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) is a multimeric E3 ligase that catalyses M1 or linear ubiquitination of activated immune receptor signalling complexes (RSCs). Mutations that disrupt linear ubiquitin assembly lead to complex disease pathologies including immunodeficiency and autoinflammation in both humans and mice, but microbial toxins that target LUBAC function have not yet been discovered. Here, we report the identification of two homologous Shigella flexneri type III secretion system effector E3 ligases IpaH1.4 and IpaH2.5, which directly interact with LUBAC subunit Heme-oxidized IRP2 ubiquitin ligase-1 (HOIL-1L) and conjugate K48-linked ubiquitin chains to the catalytic RING-between-RING domain of HOIL-1-interacting protein (HOIP). Proteasomal degradation of HOIP leads to irreversible inactivation of linear ubiquitination and blunting of NF-κB nuclear translocation in response to tumour-necrosis factor (TNF), IL-1ß and pathogen-associated molecular patterns. Loss of function studies in mammallian cells in combination with bacterial genetics explains how Shigella evades a broad spectrum of immune surveillance systems by cooperative inhibition of receptor ubiquitination and reveals the critical importance of LUBAC in host defence against pathogens.


Subject(s)
NF-kappa B/metabolism , Shigella flexneri/metabolism , Signal Transduction , Ubiquitin/metabolism , Animals , Gene Expression Regulation , Humans , Immune Evasion , Immunity, Innate , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , Tumor Necrosis Factor-alpha/immunology , Type III Secretion Systems , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
13.
Mar Pollut Bull ; 97(1-2): 294-308, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26119627

ABSTRACT

We studied short-term changes in macrozoobenthos in a 20m deep borrow pit. A boxcorer was used to sample macrobenthic infauna and a bottom sledge was used to sample macrobenthic epifauna. Sediment characteristics were determined from the boxcore samples, bed shear stress and near-bed salinity were estimated with a hydrodynamic model. Two years after the cessation of sand extraction, macrozoobenthic biomass increased fivefold in the deepest areas. Species composition changed significantly and white furrow shell (Abra alba) became abundant. Several sediment characteristics also changed significantly in the deepest parts. Macrozoobenthic species composition and biomass significantly correlated with time after cessation of sand extraction, sediment and hydrographical characteristics. Ecosystem-based landscaped sand bars were found to be effective in influencing sediment characteristics and macrozoobenthic assemblage. Significant changes in epifauna occurred in deepest parts in 2012 which coincided with the highest sedimentation rate. We recommend continuing monitoring to investigate medium and long-term impacts.


Subject(s)
Biodiversity , Environmental Monitoring/methods , Geologic Sediments , Silicon Dioxide , Animals , Biomass , Bivalvia , Ecosystem , Environment
14.
mBio ; 4(1): e00418-12, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23422410

ABSTRACT

Host cytokine responses to Brucella abortus infection are elicited predominantly by the deployment of a type IV secretion system (T4SS). However, the mechanism by which the T4SS elicits inflammation remains unknown. Here we show that translocation of the T4SS substrate VceC into host cells induces proinflammatory responses. Ectopically expressed VceC interacted with the endoplasmic reticulum (ER) chaperone BiP/Grp78 and localized to the ER of HeLa cells. ER localization of VceC required a transmembrane domain in its N terminus. Notably, the expression of VceC resulted in reorganization of ER structures. In macrophages, VceC was required for B. abortus-induced inflammation by induction of the unfolded protein response by a process requiring inositol-requiring transmembrane kinase/endonuclease 1. Altogether, these findings suggest that translocation of the T4SS effector VceC induces ER stress, which results in the induction of proinflammatory host cell responses during B. abortus infection. IMPORTANCE Brucella species are pathogens that require a type IV secretion system (T4SS) to survive in host cells and to maintain chronic infection. By as-yet-unknown pathways, the T4SS also elicits inflammatory responses in infected cells. Here we show that inflammation caused by the T4SS results in part from the sensing of a T4SS substrate, VceC, that localizes to the endoplasmic reticulum (ER), an intracellular site of Brucella replication. Possibly via binding of the ER chaperone BiP, VceC causes ER stress with concomitant expression of proinflammatory cytokines. Thus, induction of the unfolded protein response may represent a novel pathway by which host cells can detect pathogens deploying a T4SS.


Subject(s)
Bacterial Secretion Systems , Brucella abortus/metabolism , Brucella abortus/pathogenicity , Unfolded Protein Response , Virulence Factors/metabolism , Animals , Cytokines/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Chaperone BiP , Female , HeLa Cells , Humans , Inflammation Mediators/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Virulence Factors/toxicity
15.
Future Microbiol ; 7(1): 47-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22191446

ABSTRACT

Brucellosis is a global disease of domestic and wild mammals that is caused by intracellular bacteria of the genus Brucella. Although humans are not a natural reservoir for Brucella, infection in the human population is common in many countries, and brucellosis is one of the most common zoonotic infections. Brucella species have evolved to avoid the host's immune system and infection is usually characterized by long-term persistence of the bacteria. One important Brucella virulence factor for intracellular survival and persistence in the host is the type IV secretion system. This review will discuss the Brucella type IV secretion system in detail, including current knowledge of architecture and regulation, as well as the newly identified effector substrates that this system transports into host cells.


Subject(s)
Bacterial Secretion Systems , Brucella/metabolism , Brucellosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucella/genetics , Humans , Virulence Factors/genetics , Virulence Factors/metabolism
16.
Article in English | MEDLINE | ID: mdl-22919638

ABSTRACT

A large number of hypothetical genes potentially encoding small proteins of unknown function are annotated in the Brucella abortus genome. Individual deletion of 30 of these genes identified four mutants, in BAB1_0355, BAB2_0726, BAB2_0470, and BAB2_0450 that were highly attenuated for infection. BAB2_0726, an YbgT-family protein located at the 3' end of the cydAB genes encoding cytochrome bd ubiquinal oxidase, was designated cydX. A B. abortus cydX mutant lacked cytochrome bd oxidase activity, as shown by increased sensitivity to H(2)O(2), decreased acid tolerance and increased resistance to killing by respiratory inhibitors. The C terminus, but not the N terminus, of CydX was located in the periplasm, suggesting that CydX is an integral cytoplasmic membrane protein. Phenotypic analysis of the cydX mutant, therefore, suggested that CydX is required for full function of cytochrome bd oxidase, possibly via regulation of its assembly or activity.


Subject(s)
Bacterial Proteins/metabolism , Brucella abortus/enzymology , Membrane Proteins/metabolism , Oxidoreductases/metabolism , Animals , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucella abortus/metabolism , Brucellosis/microbiology , Brucellosis/pathology , Disease Models, Animal , Female , Gene Deletion , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Oxidoreductases/genetics , Virulence
17.
Science ; 337(6093): 477-81, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22722251

ABSTRACT

Defensins are antimicrobial peptides that contribute broadly to innate immunity, including protection of mucosal tissues. Human α-defensin (HD) 6 is highly expressed by secretory Paneth cells of the small intestine. However, in contrast to the other defensins, it lacks appreciable bactericidal activity. Nevertheless, we report here that HD6 affords protection against invasion by enteric bacterial pathogens in vitro and in vivo. After stochastic binding to bacterial surface proteins, HD6 undergoes ordered self-assembly to form fibrils and nanonets that surround and entangle bacteria. This self-assembly mechanism occurs in vivo, requires histidine-27, and is consistent with x-ray crystallography data. These findings support a key role for HD6 in protecting the small intestine against invasion by diverse enteric pathogens and may explain the conservation of HD6 throughout Hominidae evolution.


Subject(s)
Immunity, Innate , Immunity, Mucosal , Intestine, Small/immunology , alpha-Defensins/chemistry , alpha-Defensins/metabolism , Adhesins, Bacterial/metabolism , Animals , Bacterial Proteins/metabolism , Cell Line , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/ultrastructure , Intestine, Small/microbiology , Intestine, Small/ultrastructure , Macromolecular Substances/chemistry , Macromolecular Substances/immunology , Macromolecular Substances/metabolism , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Models, Molecular , Nanostructures , Paneth Cells/immunology , Paneth Cells/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/ultrastructure , Yersinia enterocolitica/immunology , Yersinia enterocolitica/pathogenicity , alpha-Defensins/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism
18.
PLoS One ; 4(5): e5519, 2009.
Article in English | MEDLINE | ID: mdl-19436743

ABSTRACT

Brucella ovis is a veterinary pathogen associated with epididymitis in sheep. Despite its genetic similarity to the zoonotic pathogens B. abortus, B. melitensis and B. suis, B. ovis does not cause zoonotic disease. Genomic analysis of the type strain ATCC25840 revealed a high percentage of pseudogenes and increased numbers of transposable elements compared to the zoonotic Brucella species, suggesting that genome degradation has occurred concomitant with narrowing of the host range of B. ovis. The absence of genomic island 2, encoding functions required for lipopolysaccharide biosynthesis, as well as inactivation of genes encoding urease, nutrient uptake and utilization, and outer membrane proteins may be factors contributing to the avirulence of B. ovis for humans. A 26.5 kb region of B. ovis ATCC25840 Chromosome II was absent from all the sequenced human pathogenic Brucella genomes, but was present in all of 17 B. ovis isolates tested and in three B. ceti isolates, suggesting that this DNA region may be of use for differentiating B. ovis from other Brucella spp. This is the first genomic analysis of a non-zoonotic Brucella species. The results suggest that inactivation of genes involved in nutrient acquisition and utilization, cell envelope structure and urease may have played a role in narrowing of the tissue tropism and host range of B. ovis.


Subject(s)
Brucella ovis/genetics , Genome, Bacterial , Host-Pathogen Interactions/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucella ovis/pathogenicity , DNA Transposable Elements , Gene Deletion , Sheep/microbiology
19.
J Bacteriol ; 189(10): 3922-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17351045

ABSTRACT

MisL is a Salmonella enterica serotype Typhimurium fibronectin binding protein whose expression is induced during infection of mice. T-POP transposon mutagenesis identified marT as a positive regulatory element controlling expression of a misL::lacZYA transcriptional fusion. Gel shift analysis identified MarT as a transcriptional activator of the misL promoter.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Trans-Activators/metabolism , Gene Expression Regulation, Bacterial , Lac Operon , Mutagenesis, Insertional , Promoter Regions, Genetic/physiology , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL