Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 389(25): 2355-2362, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38118023

ABSTRACT

Melioidosis, caused by Burkholderia pseudomallei, is a rare but potentially fatal bacterial disease endemic to tropical and subtropical regions worldwide. It is typically acquired through contact with contaminated soil or fresh water. Before this investigation, B. pseudomallei was not known to have been isolated from the environment in the continental United States. Here, we report on three patients living in the same Mississippi Gulf Coast county who presented with melioidosis within a 3-year period. They were infected by the same Western Hemisphere B. pseudomallei strain that was discovered in three environmental samples collected from the property of one of the patients. These findings indicate local acquisition of melioidosis from the environment in the Mississippi Gulf Coast region.


Subject(s)
Burkholderia pseudomallei , Environmental Microbiology , Melioidosis , Humans , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/isolation & purification , Melioidosis/epidemiology , Melioidosis/microbiology , United States/epidemiology
2.
EBioMedicine ; 95: 104772, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37634385

ABSTRACT

BACKGROUND: Malaria in pregnancy (MIP) causes higher morbidity in primigravid compared to multigravid women; however, the correlates and mechanisms underlying this gravidity-dependent protection remain incompletely understood. We aimed to compare the cellular immune response between primigravid and multigravid women living in a malaria-endemic region and assess for correlates of protection against MIP. METHODS: We characterised the second trimester cellular immune response among 203 primigravid and multigravid pregnant women enrolled in two clinical trials of chemoprevention in eastern Uganda, utilizing RNA sequencing, flow cytometry, and functional assays. We compared responses across gravidity and determined associations with parasitaemia during pregnancy and placental malaria. FINDINGS: Using whole blood RNA sequencing, no significant differentially expressed genes were identified between primigravid (n = 12) and multigravid (n = 11) women overall (log 2(FC) > 2, FDR < 0.1). However, primigravid (n = 49) women had higher percentages of malaria-specific, non-naïve CD4+ T cells that co-expressed IL-10 and IFNγ compared with multigravid (n = 85) women (p = 0.000023), and higher percentages of these CD4+ T cells were associated with greater risks of parasitaemia in pregnancy (Rs = 0.49, p = 0.001) and placental malaria (p = 0.0073). These IL-10 and IFNγ co-producing CD4+ T cells had a genomic signature of Tr1 cells, including expression of transcription factors cMAF and BATF and cell surface makers CTLA4 and LAG-3. INTERPRETATION: Malaria-specific Tr1 cells were highly prevalent in primigravid Ugandan women, and their presence correlated with a higher risk of malaria in pregnancy. Understanding whether suppression of Tr1 cells plays a role in naturally acquired gravidity-dependent immunity may aid the development of new vaccines or treatments for MIP. FUNDING: This work was funded by NIH (PO1 HD059454, U01 AI141308, U19 AI089674, U01 AI155325, U01 AI150741), the March of Dimes (Basil O'Connor award), and the Bill and Melinda Gates Foundation (OPP 1113682).


Subject(s)
Interleukin-10 , T-Lymphocytes, Regulatory , Pregnancy , Female , Humans , Gravidity , Placenta , CD4-Positive T-Lymphocytes
3.
Emerg Infect Dis ; 29(3): 618-621, 2023 03.
Article in English | MEDLINE | ID: mdl-36823515

ABSTRACT

Burkholderia thailandensis, an opportunistic pathogen found in the environment, is a bacterium closely related to B. pseudomallei, the cause of melioidosis. Human B. thailandensis infections are uncommon. We isolated B. thailandensis from water in Texas and Puerto Rico and soil in Mississippi in the United States, demonstrating a potential public health risk.


Subject(s)
Burkholderia Infections , Burkholderia pseudomallei , Burkholderia , Melioidosis , United States , Humans , Burkholderia Infections/microbiology
4.
Sci Transl Med ; 15(680): eadd9012, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36696483

ABSTRACT

Natural killer (NK) cells likely play an important role in immunity to malaria, but the effect of repeated malaria on NK cell responses remains unclear. Here, we comprehensively profiled the NK cell response in a cohort of 264 Ugandan children. Repeated malaria exposure was associated with expansion of an atypical, CD56neg population of NK cells that differed transcriptionally, epigenetically, and phenotypically from CD56dim NK cells, including decreased expression of PLZF and the Fc receptor γ-chain, increased histone methylation, and increased protein expression of LAG-3, KIR, and LILRB1. CD56neg NK cells were highly functional and displayed greater antibody-dependent cellular cytotoxicity than CD56dim NK cells. Higher frequencies of CD56neg NK cells were associated with protection against symptomatic malaria and high parasite densities. After marked reductions in malaria transmission, frequencies of these cells rapidly declined, suggesting that continuous exposure to Plasmodium falciparum is required to maintain this modified, adaptive-like NK cell subset.


Subject(s)
Killer Cells, Natural , Malaria , Child , Humans , CD56 Antigen/metabolism , Plasmodium falciparum , Receptors, Fc
6.
Elife ; 112022 10 14.
Article in English | MEDLINE | ID: mdl-36239699

ABSTRACT

Background: The great majority of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infections are mild and uncomplicated, but some individuals with initially mild COVID-19 progressively develop more severe symptoms. Furthermore, there is substantial heterogeneity in SARS-CoV-2-specific memory immune responses following infection. There remains a critical need to identify host immune biomarkers predictive of clinical and immunological outcomes in SARS-CoV-2-infected patients. Methods: Leveraging longitudinal samples and data from a clinical trial (N=108) in SARS-CoV-2-infected outpatients, we used host proteomics and transcriptomics to characterize the trajectory of the immune response in COVID-19 patients. We characterized the association between early immune markers and subsequent disease progression, control of viral shedding, and SARS-CoV-2-specific T cell and antibody responses measured up to 7 months after enrollment. We further compared associations between early immune markers and subsequent T cell and antibody responses following natural infection with those following mRNA vaccination. We developed machine-learning models to predict patient outcomes and validated the predictive model using data from 54 individuals enrolled in an independent clinical trial. Results: We identify early immune signatures, including plasma RIG-I levels, early IFN signaling, and related cytokines (CXCL10, MCP1, MCP-2, and MCP-3) associated with subsequent disease progression, control of viral shedding, and the SARS-CoV-2-specific T cell and antibody response measured up to 7 months after enrollment. We found that several biomarkers for immunological outcomes are shared between individuals receiving BNT162b2 (Pfizer-BioNTech) vaccine and COVID-19 patients. Finally, we demonstrate that machine-learning models using 2-7 plasma protein markers measured early within the course of infection are able to accurately predict disease progression, T cell memory, and the antibody response post-infection in a second, independent dataset. Conclusions: Early immune signatures following infection can accurately predict clinical and immunological outcomes in outpatients with COVID-19 using validated machine-learning models. Funding: Support for the study was provided from National Institute of Health/National Institute of Allergy and Infectious Diseases (NIH/NIAID) (U01 AI150741-01S1 and T32-AI052073), the Stanford's Innovative Medicines Accelerator, National Institutes of Health/National Institute on Drug Abuse (NIH/NIDA) DP1DA046089, and anonymous donors to Stanford University. Peginterferon lambda provided by Eiger BioPharmaceuticals.


Subject(s)
COVID-19 , Humans , Antibodies, Viral , Biomarkers , BNT162 Vaccine , Cytokines/metabolism , Disease Progression , RNA, Messenger , SARS-CoV-2 , Clinical Trials as Topic
7.
Cell Rep Med ; 3(6): 100640, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35588734

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific CD4+ T cells are likely important in immunity against coronavirus 2019 (COVID-19), but our understanding of CD4+ longitudinal dynamics following infection and of specific features that correlate with the maintenance of neutralizing antibodies remains limited. Here, we characterize SARS-CoV-2-specific CD4+ T cells in a longitudinal cohort of 109 COVID-19 outpatients enrolled during acute infection. The quality of the SARS-CoV-2-specific CD4+ response shifts from cells producing interferon gamma (IFNγ) to tumor necrosis factor alpha (TNF-α) from 5 days to 4 months post-enrollment, with IFNγ-IL-21-TNF-α+ CD4+ T cells the predominant population detected at later time points. Greater percentages of IFNγ-IL-21-TNF-α+ CD4+ T cells on day 28 correlate with SARS-CoV-2-neutralizing antibodies measured 7 months post-infection (⍴ = 0.4, p = 0.01). mRNA vaccination following SARS-CoV-2 infection boosts both IFNγ- and TNF-α-producing, spike-protein-specific CD4+ T cells. These data suggest that SARS-CoV-2-specific, TNF-α-producing CD4+ T cells may play an important role in antibody maintenance following COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Neutralizing , CD4-Positive T-Lymphocytes , Humans , Outpatients , T-Lymphocytes , Tumor Necrosis Factor-alpha
8.
Res Sq ; 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35132407

ABSTRACT

The great majority of SARS-CoV-2 infections are mild and uncomplicated, but some individuals with initially mild COVID-19 progressively develop more severe symptoms. Furthermore, there is substantial heterogeneity in SARS-CoV-2-specific memory immune responses following infection. There remains a critical need to identify host immune biomarkers predictive of clinical and immunologic outcomes in SARS-CoV-2-infected patients. Leveraging longitudinal samples and data from a clinical trial in SARS-CoV-2 infected outpatients, we used host proteomics and transcriptomics to characterize the trajectory of the immune response in COVID-19 patients within the first 2 weeks of symptom onset. We identify early immune signatures, including plasma RIG-I levels, early interferon signaling, and related cytokines (CXCL10, MCP1, MCP-2 and MCP-3) associated with subsequent disease progression, control of viral shedding, and the SARS-CoV-2 specific T cell and antibody response measured up to 7 months after enrollment. We found that several biomarkers for immunological outcomes are shared between individuals receiving BNT162b2 (Pfizer-BioNTech) vaccine and COVID-19 patients. Finally, we demonstrate that machine learning models using 7-10 plasma protein markers measured early within the course of infection are able to accurately predict disease progression, T cell memory, and the antibody response post-infection in a second, independent dataset.

9.
PLoS One ; 14(12): e0225588, 2019.
Article in English | MEDLINE | ID: mdl-31841511

ABSTRACT

Macrophages can reprogram their metabolism in response to the surrounding stimuli, which affects their capacity to kill intracellular pathogens. We have investigated the metabolic and immune status of human macrophages after infection with the intracellular trypanosomatid parasites Leishmania donovani, L. amazonensis and T. cruzi and their capacity to respond to a classical polarizing stimulus (LPS and IFN-γ). We found that macrophages infected with Leishmania preferentially upregulate oxidative phosphorylation, which could be contributed by both host cell and parasite, while T. cruzi infection did not significantly increase glycolysis or oxidative phosphorylation. Leishmania and T. cruzi infect macrophages without triggering a strong inflammatory cytokine response, but infection does not prevent a potent response to LPS and IFN-γ. Infection appears to prime macrophages, since the cytokine response to activation with LPS and IFN-γ is more intense in infected macrophages compared to uninfected ones. Metabolic polarization in macrophages can influence infection and immune evasion of these parasites since preventing macrophage cytokine responses would help parasites to establish a persistent infection. However, macrophages remain responsive to classical inflammatory stimuli and could still trigger inflammatory cytokine secretion by macrophages.


Subject(s)
Chagas Disease/immunology , Cytokines/metabolism , Leishmaniasis/immunology , Macrophage Activation , Macrophages/immunology , 3T3 Cells , Animals , Cells, Cultured , Chagas Disease/blood , Chagas Disease/parasitology , Cytokines/immunology , Healthy Volunteers , Humans , Leishmania donovani/immunology , Leishmania donovani/isolation & purification , Leishmania mexicana/immunology , Leishmania mexicana/isolation & purification , Leishmaniasis/blood , Leishmaniasis/parasitology , Macrophages/metabolism , Metabolome/immunology , Mice , Oxidative Phosphorylation , Primary Cell Culture , Trypanosoma cruzi/immunology , Trypanosoma cruzi/isolation & purification , Up-Regulation
10.
Immunohorizons ; 3(11): 511-518, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690559

ABSTRACT

Malaria is a highly inflammatory disease caused by the protozoan parasite Plasmodium During the blood stage of infection, patients exhibit fever with high levels of inflammatory cytokines in their blood. However, when cells of the immune system are incubated with the parasite in vitro, their cytokine response is low. In particular, human primary dendritic cells (DCs) respond to Plasmodium falciparum-infected erythrocytes by upregulating maturation markers and chemokines but lack a substantial cytokine response. Because oxidative stress is a trigger of inflammatory cytokines in malaria and synergizes with P. falciparum to induce IL-1ß secretion by macrophages, we assessed whether oxidative stress has an impact on DC maturation and function in response to P. falciparum Using xanthine oxidase, a reactive oxygen species- (ROS) producing enzyme that is increased during malaria, we observed that exposure to extracellular ROS potentiated DC maturation in response to the parasite. Xanthine oxidase-derived ROS increased parasite-induced cytokine secretion and CD80 surface expression in DCs. This enhanced maturation phenotype boosted the DCs' ability to prime autologous naive CD4+ T cells, resulting in higher T cell proliferation in vitro. Xanthine oxidase-derived ROS did not have an effect on the cytokines produced by primed T cells. We propose that oxidative stress during malaria contributes to the inflammatory response by enhancing the magnitude of DC and CD4+ T cell responses without changing the quality.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Erythrocytes/parasitology , Malaria, Falciparum/immunology , Oxidative Stress , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/parasitology , Humans , Lymphocyte Activation , Plasmodium falciparum
11.
EMBO Mol Med ; 11(8): e9903, 2019 08.
Article in English | MEDLINE | ID: mdl-31265218

ABSTRACT

Malaria is a highly inflammatory disease caused by Plasmodium infection of host erythrocytes. However, the parasite does not induce inflammatory cytokine responses in macrophages in vitro and the source of inflammation in patients remains unclear. Here, we identify oxidative stress, which is common in malaria, as an effective trigger of the inflammatory activation of macrophages. We observed that extracellular reactive oxygen species (ROS) produced by xanthine oxidase (XO), an enzyme upregulated during malaria, induce a strong inflammatory cytokine response in primary human monocyte-derived macrophages. In malaria patients, elevated plasma XO activity correlates with high levels of inflammatory cytokines and with the development of cerebral malaria. We found that incubation of macrophages with plasma from these patients can induce a XO-dependent inflammatory cytokine response, identifying a host factor as a trigger for inflammation in malaria. XO-produced ROS also increase the synthesis of pro-IL-1ß, while the parasite activates caspase-1, providing the two necessary signals for the activation of the NLRP3 inflammasome. We propose that XO-produced ROS are a key factor for the trigger of inflammation during malaria.


Subject(s)
Inflammation/enzymology , Macrophages/enzymology , Malaria, Cerebral/enzymology , Malaria, Falciparum/enzymology , Oxidative Stress , Plasmodium falciparum/pathogenicity , Reactive Oxygen Species/metabolism , Xanthine Oxidase/metabolism , Caspase 1/metabolism , Cells, Cultured , Cytokines/metabolism , Host-Pathogen Interactions , Humans , Inflammation/blood , Inflammation/parasitology , Inflammation Mediators/metabolism , Macrophage Activation , Macrophages/parasitology , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction
12.
Front Microbiol ; 9: 482, 2018.
Article in English | MEDLINE | ID: mdl-29599764

ABSTRACT

Bacillus anthracis is an endemic soil bacterium that exhibits two different lifestyles. In the soil environment, B. anthracis undergoes a cycle of saprophytic growth, sporulation, and germination. In mammalian hosts, the pathogenic lifestyle of B. anthracis is spore germination followed by vegetative cell replication, but cells do not sporulate. During infection, and in specific culture conditions, transcription of the structural genes for the anthrax toxin proteins and the biosynthetic operon for capsule synthesis is positively controlled by the regulatory protein AtxA. A critical role for the atxA gene in B. anthracis virulence has been established. Here we report an inverse relationship between toxin production and sporulation that is linked to AtxA levels. During culture in conditions favoring sporulation, B. anthracis produces little to no AtxA. When B. anthracis is cultured in conditions favoring toxin gene expression, AtxA is expressed at relatively high levels and sporulation rate and efficiency are reduced. We found that a mutation within the atxA promoter region resulting in AtxA over-expression leads to a marked sporulation defect. The sporulation phenotype of the mutant is dependent upon pXO2-0075, an atxA-regulated open reading frame located on virulence plasmid pXO2. The predicted amino acid sequence of the pXO2-0075 protein has similarity to the sensor domain of sporulation sensor histidine kinases. It was shown previously that pXO2-0075 overexpression suppresses sporulation. We have designated pXO2-0075 "skiA" for "sporulation kinase inhibitor." Our results indicate that in addition to serving as a positive regulator of virulence gene expression, AtxA modulates B. anthracis development.

13.
Proc Natl Acad Sci U S A ; 114(49): E10568-E10577, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29162686

ABSTRACT

Dendritic cells (DCs) are activated by pathogens to initiate and shape immune responses. We found that the activation of DCs by Plasmodium falciparum, the main causative agent of human malaria, induces a highly unusual phenotype by which DCs up-regulate costimulatory molecules and secretion of chemokines, but not of cytokines typical of inflammatory responses (IL-1ß, IL-6, IL-10, TNF). Similar results were obtained with DCs obtained from malaria-naïve US donors and malaria-experienced donors from Mali. Contact-dependent cross-talk between the main DC subsets, plasmacytoid and myeloid DCs (mDCs) was necessary for increased chemokine and IFN-α secretion in response to the parasite. Despite the absence of inflammatory cytokine secretion, mDCs incubated with P. falciparum-infected erythrocytes activated antigen-specific naïve CD4+ T cells to proliferate and secrete Th1-like cytokines. This unexpected response of human mDCs to P. falciparum exhibited a transcriptional program distinct from a classical LPS response, pointing to unique P. falciparum-induced activation pathways that may explain the uncharacteristic immune response to malaria.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Erythrocytes/parasitology , Host-Parasite Interactions , Lymphocyte Activation , Plasmodium falciparum/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/parasitology , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lipopolysaccharides/pharmacology , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mali , Plasmodium falciparum/growth & development , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
14.
J Clin Invest ; 126(10): 4016-4029, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27643439

ABSTRACT

Cerebral malaria is characterized by cytoadhesion of Plasmodium falciparum-infected red blood cells (Pf-iRBCs) to endothelial cells in the brain, disruption of the blood-brain barrier, and cerebral microhemorrhages. No available antimalarial drugs specifically target the endothelial disruptions underlying this complication, which is responsible for the majority of malaria-associated deaths. Here, we have demonstrated that ruptured Pf-iRBCs induce activation of ß-catenin, leading to disruption of inter-endothelial cell junctions in human brain microvascular endothelial cells (HBMECs). Inhibition of ß-catenin-induced TCF/LEF transcription in the nucleus of HBMECs prevented the disruption of endothelial junctions, confirming that ß-catenin is a key mediator of P. falciparum adverse effects on endothelial integrity. Blockade of the angiotensin II type 1 receptor (AT1) or stimulation of the type 2 receptor (AT2) abrogated Pf-iRBC-induced activation of ß-catenin and prevented the disruption of HBMEC monolayers. In a mouse model of cerebral malaria, modulation of angiotensin II receptors produced similar effects, leading to protection against cerebral malaria, reduced cerebral hemorrhages, and increased survival. In contrast, AT2-deficient mice were more susceptible to cerebral malaria. The interrelation of the ß-catenin and the angiotensin II signaling pathways opens immediate host-targeted therapeutic possibilities for cerebral malaria and other diseases in which brain endothelial integrity is compromised.


Subject(s)
Capillary Permeability , Endothelial Cells/physiology , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Receptor, Angiotensin, Type 2/metabolism , beta Catenin/physiology , Active Transport, Cell Nucleus , Antimalarials/pharmacology , Biphenyl Compounds/pharmacology , Brain/blood supply , Brain/parasitology , Cell Adhesion , Cells, Cultured , Endothelial Cells/parasitology , Endothelium, Vascular/parasitology , Endothelium, Vascular/pathology , Humans , Intercellular Junctions/metabolism , Irbesartan , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Microvessels/pathology , Plasmodium falciparum , Tetrazoles/pharmacology
15.
PLoS One ; 10(9): e0138191, 2015.
Article in English | MEDLINE | ID: mdl-26376293

ABSTRACT

Angiotensin II, a peptide hormone that regulates blood pressure, has been proposed as a protective factor against cerebral malaria based on a genetic analysis. In vitro studies have documented an inhibitory effect of angiotensin II on Plasmodium growth, while studies using chemical inhibitors of angiotensin II in mice showed protection against experimental cerebral malaria but not major effects on parasite growth. To determine whether the level of angiotensin II affects Plasmodium growth and/or disease outcome in malaria, elevated levels of angiotensin II were induced in mice by intradermal implantation of osmotic mini-pumps providing constant release of this hormone. Mice were then infected with P. berghei and monitored for parasitemia and incidence of cerebral malaria. Mice infused with angiotensin II showed decreased parasitemia seven days after infection. The development of experimental cerebral malaria was delayed and a moderate increase in survival was observed in mice with elevated angiotensin II, as confirmed by decreased number of cerebral hemorrhages compared to controls. The results presented here show for the first time the effect of elevated levels of angiotensin II in an in vivo model of malaria. The decreased pathogenesis observed in mice complements a previous human genetic study, reinforcing the hypothesis of a beneficial effect of angiotensin II in malaria.


Subject(s)
Angiotensin II/pharmacology , Disease Models, Animal , Malaria, Cerebral/prevention & control , Malaria/prevention & control , Parasitemia/prevention & control , Plasmodium berghei/physiology , Animals , Humans , Malaria/blood , Malaria/parasitology , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Parasitemia/blood , Parasitemia/parasitology , Vasoconstrictor Agents/pharmacology
16.
Infect Immun ; 83(6): 2542-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25847961

ABSTRACT

The enteric bacterium Proteus mirabilis is associated with a significant number of catheter-associated urinary tract infections (UTIs). Strict regulation of the antagonistic processes of adhesion and motility, mediated by fimbriae and flagella, respectively, is essential for disease progression. Previously, the transcriptional regulator MrpJ, which is encoded by the mrp fimbrial operon, has been shown to repress both swimming and swarming motility. Here we show that MrpJ affects an array of cellular processes beyond adherence and motility. Microarray analysis found that expression of mrpJ mimicking levels observed during UTIs leads to differential expression of 217 genes related to, among other functions, bacterial virulence, type VI secretion, and metabolism. We probed the molecular mechanism of transcriptional regulation by MrpJ using transcriptional reporters and chromatin immunoprecipitation (ChIP). Binding of MrpJ to two virulence-associated target gene promoters, the promoters of the flagellar master regulator flhDC and mrp itself, appears to be affected by the condensation state of the native chromosome, although both targets share a direct MrpJ binding site proximal to the transcriptional start. Furthermore, an mrpJ deletion mutant colonized the bladders of mice at significantly lower levels in a transurethral model of infection. Additionally, we observed that mrpJ is widely conserved in a collection of recent clinical isolates. Altogether, these findings support a role of MrpJ as a global regulator of P. mirabilis virulence.


Subject(s)
Bacterial Proteins/metabolism , Flagella/metabolism , Gene Expression Regulation, Bacterial/physiology , Operon/genetics , Proteus mirabilis/metabolism , Repressor Proteins/metabolism , Animals , Bacterial Proteins/genetics , Chromosomes, Bacterial , Mice , Mutation , Proteus mirabilis/pathogenicity , Repressor Proteins/genetics , Transcription, Genetic , Urinary Tract Infections/microbiology , Virulence
17.
Curr Rheumatol Rep ; 16(2): 401, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24390755

ABSTRACT

Malaria, which is caused by Plasmodium parasite erythrocyte infection, is a highly inflammatory disease with characteristic periodic fevers caused by the synchronous rupture of infected erythrocytes to release daughter parasites. Despite the importance of inflammation in the pathology and mortality induced by malaria, the parasite-derived factors inducing the inflammatory response are still not well characterized. Uric acid is emerging as a central inflammatory molecule in malaria. Not only is uric acid found in the precipitated form in infected erythrocytes, but high concentrations of hypoxanthine, a precursor for uric acid, also accumulate in infected erythrocytes. Both are released upon infected erythrocyte rupture into the circulation where hypoxanthine would be converted into uric acid and precipitated uric acid would encounter immune cells. Uric acid is an important contributor to inflammatory cytokine secretion, dendritic cell and T cell responses induced by Plasmodium, suggesting uric acid as a novel molecular target for anti-inflammatory therapies in malaria.


Subject(s)
Inflammation Mediators/blood , Inflammation/blood , Malaria/blood , Uric Acid/blood , Animals , Erythrocytes/metabolism , Erythrocytes/parasitology , Host-Parasite Interactions , Humans , Inflammation/parasitology , Malaria/parasitology , Plasmodium/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...