Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Commun Biol ; 6(1): 1010, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37798443

ABSTRACT

Pyroptosis is a cell death process that causes inflammation and contributes to numerous diseases. Pyroptosis is mediated by caspase-1 family proteases that cleave the pore-forming protein gasdermin D, causing plasma membrane rupture and release of pathogenic cellular contents. We previously identified muscimol as a small molecule that prevents plasma membrane rupture during pyroptosis via an unidentified mechanism. Here, we show that muscimol has reversible activity to prevent cellular lysis without affecting earlier pyroptotic events. Although muscimol is a well-characterized agonist for neuronal GABAA receptors, muscimol protection is not altered by GABAA receptor antagonists or recapitulated by other GABAA agonists, suggesting that muscimol acts via a novel mechanism. We find that muscimol blocks oligomerization of ninjurin-1, which is required for plasma membrane rupture downstream of gasdermin D pore formation. Our structure-activity relationship studies reveal distinct molecular determinants defining inhibition of pyroptotic lysis compared to GABAA binding. In addition, we demonstrate that muscimol reduces lethality during LPS-induced septic shock. Together, these findings demonstrate that ninjurin-1-mediated plasma membrane rupture can be pharmacologically modulated and pave the way toward identification of therapeutic strategies for pathologic conditions associated with pyroptosis.


Subject(s)
Gasdermins , Pyroptosis , Muscimol/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Cell Membrane/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism
3.
mBio ; 14(4): e0054023, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37306512

ABSTRACT

The cellular processes that support human coronavirus replication and contribute to the pathogenesis of severe disease remain incompletely understood. Many viruses, including coronaviruses, cause endoplasmic reticulum (ER) stress during infection. IRE1α is a component of the cellular response to ER stress that initiates non-conventional splicing of XBP1 mRNA. Spliced XBP1 encodes a transcription factor that induces the expression of ER-related targets. Activation of the IRE1α-XBP1 pathway occurs in association with risk factors for severe human coronavirus infection. In this study, we found that the human coronaviruses HCoV-OC43 (human coronavirus OC43) and SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) both robustly activate the IRE1α-XBP1 branch of the unfolded protein response in cultured cells. Using IRE1α nuclease inhibitors and genetic knockdown of IRE1α and XBP1, we found that these host factors are required for optimal replication of both viruses. Our data suggest that IRE1α supports infection downstream of initial viral attachment and entry. In addition, we found that ER stress-inducing conditions are sufficient to enhance human coronavirus replication. Furthermore, we found markedly increased XBP1 in circulation in human patients with severe coronavirus disease 2019 (COVID-19). Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection. IMPORTANCE There is a critical need to understand the cellular processes co-opted during human coronavirus replication, with an emphasis on identifying mechanisms underlying severe disease and potential therapeutic targets. Here, we demonstrate that the host proteins IRE1α and XBP1 are required for robust infection by the human coronaviruses, SARS-CoV-2 and HCoV-OC43. IRE1α and XBP1 participate in the cellular response to ER stress and are activated during conditions that predispose to severe COVID-19. We found enhanced viral replication with exogenous IRE1α activation, and evidence that this pathway is activated in humans during severe COVID-19. Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection.


Subject(s)
COVID-19 , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , SARS-CoV-2/metabolism , Unfolded Protein Response , Endoplasmic Reticulum Stress , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
4.
Methods Mol Biol ; 2641: 1-16, 2023.
Article in English | MEDLINE | ID: mdl-37074637

ABSTRACT

Pyroptosis is a regulated form of cell death that leads to inflammation and plays a role in many different diseases. Pyroptosis was initially defined by the dependence on caspase-1, a protease which is activated by innate immune signaling complexes called inflammasomes. Caspase-1 cleaves the protein gasdermin D, releasing the N-terminal pore-forming domain, which inserts into the plasma membrane. Recent studies have revealed that other gasdermin family members form plasma membrane pores, leading to lytic cell death, and the definition of pyroptosis was revised to gasdermin-dependent cell death. In this review, we discuss how the use of the term pyroptosis has changed over time, as well as currently understood molecular mechanisms leading to pyroptosis and functional consequences of this form of regulated cell death.


Subject(s)
Gasdermins , Pyroptosis , Inflammasomes/metabolism , Cell Death , Caspase 1/metabolism
5.
Brief Bioinform ; 23(6)2022 11 19.
Article in English | MEDLINE | ID: mdl-36305426

ABSTRACT

The ongoing coronavirus disease 2019 (COVID-19) pandemic has highlighted the need to better understand virus-host interactions. We developed a network-based method that expands the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-host protein interaction network and identifies host targets that modulate viral infection. To disrupt the SARS-CoV-2 interactome, we systematically probed for potent compounds that selectively target the identified host proteins with high expression in cells relevant to COVID-19. We experimentally tested seven chemical inhibitors of the identified host proteins for modulation of SARS-CoV-2 infection in human cells that express ACE2 and TMPRSS2. Inhibition of the epigenetic regulators bromodomain-containing protein 4 (BRD4) and histone deacetylase 2 (HDAC2), along with ubiquitin-specific peptidase (USP10), enhanced SARS-CoV-2 infection. Such proviral effect was observed upon treatment with compounds JQ1, vorinostat, romidepsin and spautin-1, when measured by cytopathic effect and validated by viral RNA assays, suggesting that the host proteins HDAC2, BRD4 and USP10 have antiviral functions. We observed marked differences in antiviral effects across cell lines, which may have consequences for identification of selective modulators of viral infection or potential antiviral therapeutics. While network-based approaches enable systematic identification of host targets and selective compounds that may modulate the SARS-CoV-2 interactome, further developments are warranted to increase their accuracy and cell-context specificity.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Protein Interaction Maps , Nuclear Proteins , Transcription Factors , Antiviral Agents/pharmacology , Ubiquitin Thiolesterase , Cell Cycle Proteins
6.
Methods Mol Biol ; 2543: 179-189, 2022.
Article in English | MEDLINE | ID: mdl-36087268

ABSTRACT

Pyroptosis is a highly regulated inflammatory form of cell death that plays a role in many different diseases, including cancer. Pyroptosis was initially described to be mediated by caspase-1, which is activated by innate immune signaling complexes called inflammasomes. Inflammasomes trigger caspase-dependent activation of the pore-forming protein, gasdermin D, and plasma membrane disruption. In this protocol, we describe a method to simultaneously detect two hallmarks of inflammasome-mediated pyroptosis. Using a fluorescently tagged inflammasome adaptor protein (ASC-Citrine) and membrane-impermeable nuclear dyes, we can track inflammasome formation and plasma membrane disruption over time in the same cell population.


Subject(s)
Inflammasomes , Pyroptosis , Adaptor Proteins, Signal Transducing/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
7.
Cell Death Dis ; 10(4): 326, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975978

ABSTRACT

Pyroptosis is a programmed process of proinflammatory cell death mediated by caspase-1-related proteases that cleave the pore-forming protein, gasdermin D, causing cell lysis and release of inflammatory intracellular contents. The amino acid glycine prevents pyroptotic lysis via unknown mechanisms, without affecting caspase-1 activation or pore formation. Pyroptosis plays a critical role in diverse inflammatory diseases, including sepsis. Septic lethality is prevented by glycine treatment, suggesting that glycine-mediated cytoprotection may provide therapeutic benefit. In this study, we systematically examined a panel of small molecules, structurally related to glycine, for their ability to prevent pyroptotic lysis. We found a requirement for the carboxyl group, and limited tolerance for larger amino groups and substitution of the hydrogen R group. Glycine is an agonist for the neuronal glycine receptor, which acts as a ligand-gated chloride channel. The array of cytoprotective small molecules we identified resembles that of known glycine receptor modulators. However, using genetically deficient Glrb mutant macrophages, we found that the glycine receptor is not required for pyroptotic cytoprotection. Furthermore, protection against pyroptotic lysis is independent of extracellular chloride conductance, arguing against an effect mediated by ligand-gated chloride channels. Finally, we conducted a small-scale, hypothesis-driven small-molecule screen and identified unexpected ion channel modulators that prevent pyroptotic lysis with increased potency compared to glycine. Together, these findings demonstrate that pyroptotic lysis can be pharmacologically modulated and pave the way toward identification of therapeutic strategies for pathologic conditions associated with pyroptosis.


Subject(s)
Cytoprotection/drug effects , Glycine/analogs & derivatives , Glycine/chemistry , Macrophages/drug effects , Pyroptosis/physiology , Animals , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Caspase 1/metabolism , Cell Death , Cells, Cultured , Glycine/metabolism , Ion Channels/metabolism , Ion Channels/physiology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Receptors, Glycine/agonists , Receptors, Glycine/antagonists & inhibitors , Receptors, Glycine/metabolism , Salmonella
8.
Curr Protoc Immunol ; 122(1): e52, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30028908

ABSTRACT

Pyroptosis is a form of programmed pro-inflammatory cell death that plays a protective role in the host response to infection, but can also promote pathogenic inflammation. Pyroptosis is mediated by the cysteine protease, caspase-1. Caspase-1 cleaves gasdermin D, releasing the N-terminal pore-forming domain, which inserts into the plasma membrane and drives osmotic lysis. Caspase-1 also proteolytically activates the inflammatory cytokines interleukin 1ß (IL-1ß) and IL-18. This unit describes methods for stimulating pyroptosis and assessing subsequent loss of plasma membrane integrity. We also describe an ELISA to quantify released IL-1ß. These methods can be applied to many different types of experiments. © 2018 by John Wiley & Sons, Inc.

9.
J Vis Exp ; (135)2018 05 21.
Article in English | MEDLINE | ID: mdl-29863661

ABSTRACT

Inflammasomes are innate immune signaling platforms that are required for the successful control of many pathogenic organisms, but also promote inflammatory and autoinflammatory diseases. Inflammasomes are activated by cytosolic pattern recognition receptors, including members of the NOD-like receptor (NLR) family. These receptors oligomerize upon the detection of microbial or damage-associated stimuli. Subsequent recruitment of the adaptor protein ASC forms a microscopically visible inflammasome complex, which activates caspase-1 through proximity-induced auto-activation. Following the activation, caspase-1 cleaves pro-IL-1ß and pro-IL-18, leading to the activation and secretion of these pro-inflammatory cytokines. Caspase-1 also mediates the inflammatory form of cell death termed pyroptosis, which features the loss of membrane integrity and cell lysis. Caspase-1 cleaves gasdermin D, releasing the N-terminal fragment which forms plasma membrane pores, leading to osmotic lysis. In vitro, the activation of caspase-1 can be determined by labeling bone marrow-derived macrophages with the caspase-1 activity probe FAM-YVAD-FMK and by labeling the cells with antibodies against the adaptor protein ASC. This technique allows the identification of inflammasome formation and caspase-1 activation in individual cells using fluorescence microscopy. Pyroptotic cell death can be detected by measuring the release of cytosolic lactate dehydrogenase into the medium. This procedure is simple, cost effective and performed in a 96-well plate format, allowing adaptation for screening. In this manuscript, we show that activation of the NLRP3 inflammasome by nigericin leads to the co-localization of the adaptor protein ASC and active caspase-1, leading to pyroptosis.


Subject(s)
Inflammasomes/immunology , Macrophages/metabolism , Pyroptosis/genetics , Animals , Immunity, Innate , Mice , Signal Transduction
10.
Cell Host Microbe ; 14(2): 159-70, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23954155

ABSTRACT

Eradication of persistent intracellular bacterial pathogens with antibiotic therapy is often slow or incomplete. However, strategies to augment antibiotics are hampered by our poor understanding of the nutritional environment that sustains chronic infection. Here we show that the intracellular pathogen Brucella abortus survives and replicates preferentially in alternatively activated macrophages (AAMs), which are more abundant during chronic infection. A metabolic shift induced by peroxisome proliferator-activated receptor γ (PPARγ), which increases intracellular glucose availability, is identified as a causal mechanism promoting enhanced bacterial survival in AAMs. Glucose uptake was crucial for increased replication of B. abortus in AAMs, and for chronic infection, as inactivation of the bacterial glucose transporter gluP reduced both intracellular survival in AAMs and persistence in mice. Thus, a shift in intracellular nutrient availability induced by PPARγ promotes chronic persistence of B. abortus within AAMs, and targeting this pathway may aid in eradicating chronic infection.


Subject(s)
Brucella abortus/physiology , Glucose/metabolism , Macrophage Activation , Macrophages/microbiology , Microbial Viability , PPAR gamma/metabolism , Animals , Brucella abortus/growth & development , Brucella abortus/immunology , Brucella abortus/metabolism , Macrophages/immunology , Mice
11.
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
12.
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
13.
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
14.
J Immunol ; 187(5): 2748-54, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21804020

ABSTRACT

Activation of caspase-1 leads to pyroptosis, a program of cell death characterized by cell lysis and inflammatory cytokine release. Caspase-1 activation triggered by multiple nucleotide-binding oligomerization domain-like receptors (NLRs; NLRC4, NLRP1b, or NLRP3) leads to loss of lysosomes via their fusion with the cell surface, or lysosome exocytosis. Active caspase-1 increased cellular membrane permeability and intracellular calcium levels, which facilitated lysosome exocytosis and release of host antimicrobial factors and microbial products. Lysosome exocytosis has been proposed to mediate secretion of IL-1ß and IL-18; however, blocking lysosome exocytosis did not alter cytokine processing or release. These studies indicate two conserved secretion pathways are initiated by caspase-1, lysosome exocytosis, and a parallel pathway resulting in cytokine release, and both enhance the antimicrobial nature of pyroptosis.


Subject(s)
Apoptosis/physiology , Caspase 1/metabolism , Cytokines/metabolism , Exocytosis/physiology , Lysosomes/metabolism , Macrophages/metabolism , Animals , Blotting, Western , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Infect Immun ; 79(4): 1706-17, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300772

ABSTRACT

Brucella ovis is a major cause of reproductive failure in sheep, which is associated with epididymitis and infertility in rams. Importantly, B. ovis is one of the few Brucella species that is not zoonotic. Due to the scarcity of studies on B. ovis infection, a murine model of infection was developed. The roles of B. ovis genes encoding a putative hemagglutinin and an ABC transporter were investigated in the mouse model. The kinetics of B. ovis infection were similar in BALB/c and C57BL/6 mice, and both strains of mice developed multifocal microgranulomas in the liver and spleen, but only minimal colonization and histopathological changes were observed in the genital tract. Therefore, the mouse was considered a suitable infection model for B. ovis but not for B. ovis-induced genital disease. Two mutant strains were generated in this study (the ΔabcAB and Δhmg strains). The B. ovis ΔabcAB strain was attenuated in the spleens and livers of BALB/c mice compared to the wild-type (WT) strain (P < 0.001). Conversely, the Δhmg strain infected mice at the same level as WT B. ovis, suggesting that a putative hemagglutinin is not required for B. ovis pathogenesis. Additionally, the ΔabcAB strain did not survive in peritoneal macrophages, extracellularly in the peritoneal cavity, or in RAW 264.7 macrophages. Moreover, infection with the ΔabcAB strain was not lethal for male regulatory factor 1-knockout mice, whereas infection with the B. ovis WT strain was 100% lethal within 14 days postinfection. These results confirm that the predicted ABC transporter is required for the full virulence and survival of B. ovis in vivo.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Brucella ovis/genetics , Brucella ovis/pathogenicity , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucellosis/genetics , Brucellosis/metabolism , Brucellosis/pathology , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Virulence Factors/genetics
16.
Infect Immun ; 77(10): 4197-208, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651862

ABSTRACT

Human brucellosis is caused mainly by Brucella melitensis, which is often acquired by ingesting contaminated goat or sheep milk and cheese. Bacterial factors required for food-borne infection of humans by B. melitensis are poorly understood. In this study, a mouse model of oral infection was characterized to assess the roles of urease, the VirB type IV secretion system, and lipopolysaccharide for establishing infection through the digestive tract. B. melitensis strain 16M was consistently recovered from the mesenteric lymph node (MLN), spleen, and liver beginning at 3 or 7 day postinfection (dpi). In the gut, persistence of the inoculum was observed up to 21 dpi. No inflammatory lesions were observed in the ileum or colon during infection. Mutant strains lacking the ureABC genes of the ure1 operon, virB2, or pmm encoding phosphomannomutase were constructed and compared to the wild-type strain for infectivity through the digestive tract. Mutants lacking the virB2 and pmm genes were attenuated in the spleen (P < 0.05) and MLN (P < 0.001), respectively. The wild-type and mutant strains had similar levels of resistance to low pH and 5 or 10% bile, suggesting that the reduced colonization of mutants was not the result of reduced resistance to acid pH or bile salts. In an in vitro lymphoepithelial cell (M-cell) model, B. melitensis transited rapidly through polarized enterocyte monolayers containing M-like cells; however, transit through monolayers containing only enterocytes was reduced or absent. These results indicate that B. melitensis is able to spread systemically from the digestive tract after infection, most likely through M cells of the mucosa-associated lymphoid tissue.


Subject(s)
Bacterial Proteins/physiology , Brucella melitensis/pathogenicity , Gastrointestinal Tract/microbiology , Lipopolysaccharides/physiology , Membrane Transport Proteins/physiology , O Antigens/physiology , Urease/physiology , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Cell Line , Colon/pathology , Colony Count, Microbial , Enterocytes/microbiology , Female , Gene Deletion , Ileum/pathology , Lipopolysaccharides/genetics , Liver/microbiology , Lymph Nodes/microbiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , O Antigens/genetics , Spleen/microbiology , Urease/genetics , Virulence Factors/genetics
17.
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
18.
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
19.
Clin Vaccine Immunol ; 15(2): 208-14, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077620

ABSTRACT

The Brucella species type IV secretion system, encoded by the virB1-12 locus, is required for intracellular replication and persistent infection in vivo. The requirement of VirB proteins for infection suggests that they are expressed in vivo and may therefore represent serological markers of infection. To test this idea, we purified recombinant VirB1, VirB5, VirB11, and VirB12 and tested for their recognition by antibodies in sera from experimentally infected mice and goats by using an indirect enzyme-linked immunosorbent assay. Antibody responses to VirB12 but not to VirB1, VirB5, or VirB11 were detected in 20/20 mice experimentally inoculated with Brucella abortus and 12/12 goats experimentally infected with Brucella melitensis. The potential use of VirB12 as a serological tool for the diagnosis of brucellosis was evaluated in the natural bovine host. Serum samples from 145 cattle of known serology (29% negative and 71% positive) were analyzed for the production of antibody responses to VirB12. One hundred two cattle samples (70.3%) were positive for antibodies to VirB12, while 43 samples were negative (29.7%). A positive serological response to VirB12 correlated with positive serology to whole B. abortus antigen in 99% of samples tested. These results show that VirB12 is expressed during infection of both experimental and natural hosts of Brucella species, and they suggest that VirB12 may be a useful serodiagnostic marker for brucellosis.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial , Brucellosis, Bovine/diagnosis , Brucellosis/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Biomarkers , Brucella abortus/immunology , Brucella melitensis/immunology , Cattle , Female , Goats , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
20.
Vaccine ; 24(24): 5169-77, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16697090

ABSTRACT

Pregnant goats were employed to assess unmarked deletion mutant vaccine candidates BMDeltaasp24, BMDeltacydBA, and BMDeltavirB2, as the target host species naturally infected with Brucella melitensis. Goats were assessed for the degree of pathology associated with the vaccine strains as well as the protective immunity afforded by each strain against abortion and infection after challenge with wild-type Brucella melitensis 16M. Both BMDeltaasp24 and BMDeltavirB2 were considered safe vaccine candidates in the pregnant goat model because they did not cause abortion or colonize fetal tissues. BMDeltaasp24 was isolated from the maternal tissues only, indicating a slower rate of clearance of the vaccine strain than for BMDeltavirB2, which was not isolated from any maternal or fetal tissues. Both strains were protective against abortion and against infection in the majority of pregnant goats, although BMDeltaasp24 was more efficacious than BMDeltavirB2 against challenge infection.


Subject(s)
Brucella Vaccine/immunology , Brucella melitensis/immunology , Brucellosis/prevention & control , Vaccines, Synthetic/immunology , Animals , Brucella Vaccine/adverse effects , Disease Models, Animal , Female , Goats , Mutation , Pregnancy , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL