Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Publication year range
1.
Nature ; 626(7999): 661-669, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267581

ABSTRACT

Organisms determine the transcription rates of thousands of genes through a few modes of regulation that recur across the genome1. In bacteria, the relationship between the regulatory architecture of a gene and its expression is well understood for individual model gene circuits2,3. However, a broader perspective of these dynamics at the genome scale is lacking, in part because bacterial transcriptomics has hitherto captured only a static snapshot of expression averaged across millions of cells4. As a result, the full diversity of gene expression dynamics and their relation to regulatory architecture remains unknown. Here we present a novel genome-wide classification of regulatory modes based on the transcriptional response of each gene to its own replication, which we term the transcription-replication interaction profile (TRIP). Analysing single-bacterium RNA-sequencing data, we found that the response to the universal perturbation of chromosomal replication integrates biological regulatory factors with biophysical molecular events on the chromosome to reveal the local regulatory context of a gene. Whereas the TRIPs of many genes conform to a gene dosage-dependent pattern, others diverge in distinct ways, and this is shaped by factors such as intra-operon position and repression state. By revealing the underlying mechanistic drivers of gene expression heterogeneity, this work provides a quantitative, biophysical framework for modelling replication-dependent expression dynamics.


Subject(s)
Bacteria , DNA Replication , Gene Expression Regulation, Bacterial , Genome, Bacterial , Transcription, Genetic , Bacteria/genetics , DNA Replication/genetics , Gene Dosage/genetics , Gene Regulatory Networks , Genome, Bacterial/genetics , Operon/genetics , Sequence Analysis, RNA , Transcription, Genetic/genetics , Chromosomes, Bacterial/genetics
2.
Nature ; 579(7798): 260-264, 2020 03.
Article in English | MEDLINE | ID: mdl-32132711

ABSTRACT

The production of pore-forming toxins that disrupt the plasma membrane of host cells is a common virulence strategy for bacterial pathogens such as methicillin-resistant Staphylococcus aureus (MRSA)1-3. It is unclear, however, whether host species possess innate immune mechanisms that can neutralize pore-forming toxins during infection. We previously showed that the autophagy protein ATG16L1 is necessary for protection against MRSA strains encoding α-toxin4-a pore-forming toxin that binds the metalloprotease ADAM10 on the surface of a broad range of target cells and tissues2,5,6. Autophagy typically involves the targeting of cytosolic material to the lysosome for degradation. Here we demonstrate that ATG16L1 and other ATG proteins mediate protection against α-toxin through the release of ADAM10 on exosomes-extracellular vesicles of endosomal origin. Bacterial DNA and CpG DNA induce the secretion of ADAM10-bearing exosomes from human cells as well as in mice. Transferred exosomes protect host cells in vitro by serving as scavengers that can bind multiple toxins, and improve the survival of mice infected with MRSA in vivo. These findings indicate that ATG proteins mediate a previously unknown form of defence in response to infection, facilitating the release of exosomes that serve as decoys for bacterially produced toxins.


Subject(s)
Autophagy-Related Proteins/metabolism , Bacterial Toxins/metabolism , Exosomes/metabolism , A549 Cells , ADAM10 Protein/metabolism , Animals , Bacterial Toxins/pharmacology , Cell Survival/drug effects , DNA, Bacterial/pharmacology , Exosomes/drug effects , Exosomes/ultrastructure , Female , HEK293 Cells , Humans , Male , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Mice, Inbred C57BL , Staphylococcal Infections/mortality
3.
PLoS Pathog ; 19(9): e1011647, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37738244

ABSTRACT

The bacterial microbiota promotes the life cycle of the intestine-dwelling whipworm Trichuris by mediating hatching of parasite eggs ingested by the mammalian host. Despite the enormous disease burden associated with Trichuris colonization, the mechanisms underlying this transkingdom interaction have been obscure. Here, we used a multiscale microscopy approach to define the structural events associated with bacteria-mediated hatching of eggs for the murine model parasite Trichuris muris. Through the combination of scanning electron microscopy (SEM) and serial block face SEM (SBFSEM), we visualized the outer surface morphology of the shell and generated 3D structures of the egg and larva during the hatching process. These images revealed that exposure to hatching-inducing bacteria catalyzed asymmetric degradation of the polar plugs prior to exit by the larva. Unrelated bacteria induced similar loss of electron density and dissolution of the structural integrity of the plugs. Egg hatching was most efficient when high densities of bacteria were bound to the poles. Consistent with the ability of taxonomically distant bacteria to induce hatching, additional results suggest chitinase released from larva within the eggs degrade the plugs from the inside instead of enzymes produced by bacteria in the external environment. These findings define at ultrastructure resolution the evolutionary adaptation of a parasite for the microbe-rich environment of the mammalian gut.


Subject(s)
Microbiota , Trichuris , Mice , Animals , Microscopy, Electron, Scanning , Bacteria , Larva , Ovum , Mammals
4.
PLoS Biol ; 20(9): e3001754, 2022 09.
Article in English | MEDLINE | ID: mdl-36099266

ABSTRACT

Extracellular vesicles of endosomal origin, exosomes, mediate intercellular communication by transporting substrates with a variety of functions related to tissue homeostasis and disease. Their diagnostic and therapeutic potential has been recognized for diseases such as cancer in which signaling defects are prominent. However, it is unclear to what extent exosomes and their cargo inform the progression of infectious diseases. We recently defined a subset of exosomes termed defensosomes that are mobilized during bacterial infection in a manner dependent on autophagy proteins. Through incorporating protein receptors on their surface, defensosomes mediated host defense by binding and inhibiting pore-forming toxins secreted by bacterial pathogens. Given this capacity to serve as decoys that interfere with surface protein interactions, we investigated the role of defensosomes during infection by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent of Coronavirus Disease 2019 (COVID-19). Consistent with a protective function, exosomes containing high levels of the viral receptor ACE2 in bronchoalveolar lavage fluid (BALF) from critically ill COVID-19 patients was associated with reduced intensive care unit (ICU) and hospitalization times. We found ACE2+ exosomes were induced by SARS-CoV-2 infection and activation of viral sensors in cell culture, which required the autophagy protein ATG16L1, defining these as defensosomes. We further demonstrate that ACE2+ defensosomes directly bind and block viral entry. These findings suggest that defensosomes may contribute to the antiviral response against SARS-CoV-2 and expand our knowledge on the regulation and effects of extracellular vesicles during infection.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , Humans , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus , SARS-CoV-2
5.
J Immunol ; 211(5): 836-843, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37466391

ABSTRACT

Our previous studies identified a population of stem cell-like proliferating myeloid cells within inflamed tissues that could serve as a reservoir for tissue macrophages to adopt different activation states depending on the microenvironment. By lineage-tracing cells derived from CX3CR1+ precursors in mice during infection and profiling by single-cell RNA sequencing, in this study, we identify a cluster of BIRC5+ myeloid cells that expanded in the liver during chronic infection with either the parasite Schistosoma mansoni or the bacterial pathogen Staphylococcus aureus. In the absence of tissue-damaging toxins, S. aureus infection does not elicit these BIRC5+ cells. Moreover, deletion of BIRC5 from CX3CR1-expressing cells results in improved survival during S. aureus infection. Hence the combination of single-cell RNA sequencing and genetic fate-mapping CX3CR1+ cells revealed a toxin-dependent pathogenic role for BIRC5 in myeloid cells during S. aureus infection.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Mice , Animals , Myeloid Cells/pathology , Single-Cell Analysis , Staphylococcal Infections/microbiology
6.
Proc Natl Acad Sci U S A ; 119(31): e2123017119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881802

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen and chief among bloodstream-infecting bacteria. S. aureus produces an array of human-specific virulence factors that may contribute to immune suppression. Here, we defined the response of primary human phagocytes following infection with S. aureus using RNA-sequencing (RNA-Seq). We found that the overall transcriptional response to S. aureus was weak both in the number of genes and in the magnitude of response. Using an ex vivo bacteremia model with fresh human blood, we uncovered that infection with S. aureus resulted in the down-regulation of genes related to innate immune response and cytokine and chemokine signaling. This muted transcriptional response was conserved across diverse S. aureus clones but absent in blood exposed to heat-killed S. aureus or blood infected with the less virulent staphylococcal species Staphylococcus epidermidis. Notably, this signature was also present in patients with S. aureus bacteremia. We identified the master regulator S. aureus exoprotein expression (SaeRS) and the SaeRS-regulated pore-forming toxins as key mediators of the transcriptional suppression. The S. aureus-mediated suppression of chemokine and cytokine transcription was reflected by circulating protein levels in the plasma. Wild-type S. aureus elicited a soluble milieu that was restrictive in the recruitment of human neutrophils compared with strains lacking saeRS. Thus, S. aureus blunts the inflammatory response resulting in impaired neutrophil recruitment, which could promote the survival of the pathogen during invasive infection.


Subject(s)
Host-Pathogen Interactions , Neutrophils , Staphylococcal Infections , Staphylococcus aureus , Bacteremia/immunology , Bacteremia/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytokines/metabolism , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions/immunology , Humans , Neutrophils/immunology , Neutrophils/microbiology , Pore Forming Cytotoxic Proteins/genetics , Staphylococcal Infections/blood , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Staphylococcus epidermidis/pathogenicity , Transcription Factors/genetics , Transcription Factors/metabolism
7.
J Biol Chem ; 299(12): 105321, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37802313

ABSTRACT

Staphylococcus aureus (S. aureus) is a serious global pathogen that causes a diverse range of invasive diseases. S. aureus utilizes a family of pore-forming toxins, known as bi-component leukocidins, to evade the host immune response and promote infection. Among these is LukAB (leukocidin A/leukocidin B), a toxin that assembles into an octameric ß-barrel pore in the target cell membrane, resulting in host cell death. The established cellular receptor for LukAB is CD11b of the Mac-1 complex. Here, we show that hydrogen voltage-gated channel 1 is also required for the cytotoxicity of all major LukAB variants. We demonstrate that while each receptor is sufficient to recruit LukAB to the plasma membrane, both receptors are required for maximal lytic activity. Why LukAB requires two receptors, and how each of these receptors contributes to pore-formation remains unknown. To begin to resolve this, we performed an alanine scanning mutagenesis screen to identify mutations that allow LukAB to maintain cytotoxicity without CD11b. We discovered 30 mutations primarily localized in the stem domains of LukA and LukB that enable LukAB to exhibit full cytotoxicity in the absence of CD11b. Using crosslinking, electron microscopy, and hydroxyl radical protein footprinting, we show these mutations increase the solvent accessibility of the stem domain, priming LukAB for oligomerization. Together, our data support a model in which CD11b binding unlatches the membrane penetrating stem domains of LukAB, and this change in flexibility promotes toxin oligomerization.


Subject(s)
Bacterial Proteins , Leukocidins , Staphylococcus aureus , Toxins, Biological , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Leukocidins/genetics , Leukocidins/metabolism , Leukocidins/toxicity , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Toxins, Biological/metabolism , Mutation , Protein Binding/genetics , Protein Domains , Cell Line , CHO Cells , Cricetulus , Animals
8.
Infect Immun ; 92(2): e0052623, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38235972

ABSTRACT

Staphylococcus aureus is a gram-positive pathogen that poses a major health concern, in part due to its large array of virulence factors that allow infection and evasion of the immune system. One of these virulence factors is the bicomponent pore-forming leukocidin LukAB. The regulation of lukAB expression is not completely understood, especially in the presence of immune cells such as human polymorphonuclear neutrophils (hPMNs). Here, we screened for transcriptional regulators of lukAB during the infection of primary hPMNs. We uncovered that PerR, a peroxide sensor, is vital for hPMN-mediated induction of lukAB and that PerR upregulates cytotoxicity during the infection of hPMNs. Exposure of S. aureus to hydrogen peroxide (H2O2) alone also results in increased lukAB promoter activity, a phenotype dependent on PerR. Collectively, our data suggest that S. aureus uses PerR to sense the H2O2 produced by hPMNs to stimulate the expression of lukAB, allowing the bacteria to withstand these critical innate immune cells.IMPORTANCEStaphylococcus aureus utilizes a diverse set of virulence factors, such as leukocidins, to subvert human neutrophils, but how these toxins are regulated is incompletely defined. Here, we identified the peroxide-sensitive repressor, PerR, as a required protein involved in the induction of lukAB in the presence of primary human neutrophils, a phenotype directly linked to the ability of PerR to sense H2O2. Thus, we show that S. aureus coordinates sensing and resistance to oxidative stress with toxin production to promote pathogen survival.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Neutrophils , Hydrogen Peroxide/toxicity , Hydrogen Peroxide/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Bacterial Proteins/metabolism , Leukocidins , Staphylococcal Infections/microbiology
9.
Nat Chem Biol ; 18(7): 706-712, 2022 07.
Article in English | MEDLINE | ID: mdl-35361990

ABSTRACT

Membrane protein efflux pumps confer antibiotic resistance by extruding structurally distinct compounds and lowering their intracellular concentration. Yet, there are no clinically approved drugs to inhibit efflux pumps, which would potentiate the efficacy of existing antibiotics rendered ineffective by drug efflux. Here we identified synthetic antigen-binding fragments (Fabs) that inhibit the quinolone transporter NorA from methicillin-resistant Staphylococcus aureus (MRSA). Structures of two NorA-Fab complexes determined using cryo-electron microscopy reveal a Fab loop deeply inserted in the substrate-binding pocket of NorA. An arginine residue on this loop interacts with two neighboring aspartate and glutamate residues essential for NorA-mediated antibiotic resistance in MRSA. Peptide mimics of the Fab loop inhibit NorA with submicromolar potency and ablate MRSA growth in combination with the antibiotic norfloxacin. These findings establish a class of peptide inhibitors that block antibiotic efflux in MRSA by targeting indispensable residues in NorA without the need for membrane permeability.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Humans , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/pharmacology , Staphylococcus aureus/metabolism
10.
Infect Immun ; 91(4): e0004623, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36975788

ABSTRACT

The regulation of membrane protein activity for cellular functions is critically dependent on the composition of phospholipid membranes. Cardiolipin, a unique phospholipid found in bacterial membranes and mitochondrial membranes of eukaryotes, plays a crucial role in stabilizing membrane proteins and maintaining their function. In the human pathogen Staphylococcus aureus, the SaeRS two-component system (TCS) controls the expression of key virulence factors essential for the bacterium's virulence. The SaeS sensor kinase activates the SaeR response regulator via phosphoryl transfer to bind its gene target promoters. In this study, we report that cardiolipin is critical for sustaining the full activity of SaeRS and other TCSs in S. aureus. The sensor kinase protein SaeS binds directly to cardiolipin and phosphatidylglycerol, enabling SaeS activity. Elimination of cardiolipin from the membrane reduces SaeS kinase activity, indicating that bacterial cardiolipin is necessary for modulating the kinase activities of SaeS and other sensor kinases during infection. Moreover, the deletion of cardiolipin synthase genes cls1 and cls2 leads to reduced cytotoxicity to human neutrophils and lower virulence in a mouse model of infection. These findings suggest a model where cardiolipin modulates the kinase activity of SaeS and other sensor kinases after infection to adapt to the hostile environment of the host and expand our knowledge of how phospholipids contribute to membrane protein function.


Subject(s)
Cardiolipins , Transcription Factors , Animals , Mice , Humans , Cardiolipins/metabolism , Transcription Factors/genetics , Staphylococcus aureus , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Gene Expression Regulation, Bacterial
11.
Infect Immun ; 91(4): e0053222, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36939325

ABSTRACT

Staphylococcus aureus is a successful pathogen that produces a wide range of virulence factors that it uses to subvert and suppress the immune system. These include the bicomponent pore-forming leukocidins. How the expression of these toxins is regulated is not completely understood. Here, we describe a screen to identify transcription factors involved in the regulation of leukocidins. The most prominent discovery from this screen is that SarS, a known transcription factor which had previously been described as a repressor of alpha-toxin expression, was found to be a potent repressor of leukocidins LukED and LukSF-PV. We found that inactivating sarS resulted in increased virulence both in an ex vivo model using primary human neutrophils and in an in vivo infection model in mice. Further experimentation revealed that SarS represses leukocidins by serving as an activator of Rot, a critical repressor of toxins, as well as by directly binding and repressing the leukocidin promoters. By studying contemporary clinical isolates, we identified naturally occurring mutations in the sarS promoter that resulted in overexpression of sarS and increased repression of leukocidins in USA300 bloodstream clinical isolates. Overall, these data establish SarS as an important repressor of leukocidins and expand our understanding of how these virulence factors are being regulated in vitro and in vivo by S. aureus.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Humans , Mice , Exotoxins/genetics , Exotoxins/metabolism , Leukocidins/genetics , Neutrophils , Transcription Factors/metabolism , Virulence Factors/metabolism
12.
Microbiology (Reading) ; 169(9)2023 09.
Article in English | MEDLINE | ID: mdl-37702594

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins, produced by numerous Gram-positive pathogens. CDCs depend on host membrane cholesterol for pore formation; some CDCs also require surface-associated human CD59 (hCD59) for binding, conferring specificity for human cells. We purified a recombinant version of a putative CDC encoded in the genome of Streptococcus oralis subsp. tigurinus, tigurilysin (TGY), and used CRISPR/Cas9 to construct hCD59 knockout (KO) HeLa and JEG-3 cell lines. Cell viability assays with TGY on wild-type and hCD59 KO cells showed that TGY is a hCD59-dependent CDC. Two variants of TGY exist among S. oralis subsp. tigurinus genomes, only one of which is functional. We discovered that a single amino acid change between these two TGY variants determines its activity. Flow cytometry and oligomerization Western blots revealed that the single amino acid difference between the two TGY isoforms disrupts host cell binding and oligomerization. Furthermore, experiments with hCD59 KO cells and cholesterol-depleted cells demonstrated that TGY is fully dependent on both hCD59 and cholesterol for activity, unlike other known hCD59-dependent CDCs. Using full-length CDCs and toxin constructs differing only in the binding domain, we determined that having hCD59 dependence leads to increased lysis efficiency, conferring a potential advantage to organisms producing hCD59-dependent CDCs.


Subject(s)
Cytotoxins , Host Specificity , Humans , Cell Line, Tumor , Cytotoxins/genetics , Cholesterol , Amino Acids , CD59 Antigens/genetics
13.
J Infect Dis ; 225(8): 1460-1470, 2022 04 19.
Article in English | MEDLINE | ID: mdl-33895843

ABSTRACT

Vaccines against Staphylococcus aureus have eluded researchers for >3 decades while the burden of staphylococcal diseases has increased. Early vaccine attempts mainly used rodents to characterize preclinical efficacy, and all subsequently failed in human clinical efficacy trials. More recently, leukocidin AB (LukAB) has gained interest as a vaccine antigen. We developed a minipig deep surgical wound infection model offering 3 independent efficacy readouts: bacterial load at the superficial and at the deep-seated surgical site, and dissemination of bacteria. Due to similarities with humans, minipigs are an attractive option to study novel vaccine candidates. With this model, we characterized the efficacy of a LukAB toxoid as vaccine candidate. Compared to control animals, a 3-log reduction of bacteria at the deep-seated surgical site was observed in LukAB-treated minipigs and dissemination of bacteria was dramatically reduced. Therefore, LukAB toxoids may be a useful addition to S. aureus vaccines and warrant further study.


Subject(s)
Staphylococcal Infections , Staphylococcal Vaccines , Animals , Bacterial Load , Bacterial Proteins , Leukocidins , Staphylococcal Infections/microbiology , Staphylococcus aureus , Surgical Wound Infection/prevention & control , Swine , Swine, Miniature , Vaccination
14.
Infect Immun ; 90(4): e0005622, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35258336

ABSTRACT

Infections caused by the Gram-positive bacterium Staphylococcus aureus remain a significant health threat globally. The production of bicomponent pore-forming leukocidins plays an important role in S. aureus pathogenesis. Transcriptionally, these toxins are primarily regulated by the Sae and Agr regulatory systems. However, the posttranslational regulation of these toxins is largely unexplored. In particular, one of the leukocidins, LukAB, has been shown to be both secreted into the extracellular milieu and associated with the bacterial cell envelope. Here, we report that a major cell wall hydrolase, autolysin (Atl), controls the sorting of LukAB from the cell envelope to the extracellular milieu, an effect independent of transcriptional regulation. By influencing the sorting of LukAB, Atl modulates S. aureus cytotoxicity toward primary human neutrophils. Mechanistically, we found that the reduction in peptidoglycan cleavage and increased LukAB secretion in the atl mutant can be reversed through the supplementation of exogenous mutanolysin. Altogether, our study revealed that the cell wall hydrolase activity of Atl and the cleavage of peptidoglycan play an important role in controlling the sorting of S. aureus toxins during secretion.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Bacterial Proteins/genetics , Bacterial Proteins/toxicity , Humans , Leukocidins , N-Acetylmuramoyl-L-alanine Amidase/genetics , Peptidoglycan , Staphylococcal Infections/microbiology , Virulence
15.
Antimicrob Agents Chemother ; 66(1): e0155921, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34748383

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) bloodstream infections are associated with significant morbidity and mortality. MRSA secretes a number of virulence factors and pore-forming toxins that enable tissue invasion. Prior studies have found associations between decreased toxin production and poor outcomes in invasive MRSA infection, particularly in pneumonia. In this retrospective observational cohort study of MRSA bacteremia in adult patients from 2007 to 2015, we examined whether cytotoxicity was associated with 30-day mortality. Isolates were obtained from 776 patients and screened for cytotoxicity in a human HL-60 cell model, antimicrobial susceptibility, and spa type, and clinical data were abstracted from charts. We did not find an association between low cytotoxic activity and 30-day mortality in univariate logistic regression analyses. There was a difference in distribution of the genotypes across cytotoxicity phenotypes, with spa-CC008 accounting for a larger proportion of isolates in the high cytotoxicity group. Isolates with a skin and soft tissue primary infective site had a higher median cytotoxicity. There was no association between cytotoxicity and host factors such as age or comorbidity burden. The isolates in our study came from heterogeneous primary sites of infection and were predominantly from spa-CC002 and spa-CC008 lineages, so it is possible that findings in prior studies reflect a different distribution in genotypes and clinical syndromes. Overall, in this large study of cytotoxicity of MRSA bloodstream isolates, we did not find the low cytotoxicity phenotype to be predictive of poor outcomes in MRSA bacteremia.


Subject(s)
Bacteremia , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Retrospective Studies , Staphylococcal Infections/drug therapy , Virulence Factors/genetics
16.
Blood ; 135(26): 2388-2401, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32232483

ABSTRACT

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Subject(s)
Graft vs Host Disease/prevention & control , Intestinal Diseases/prevention & control , Organoids , T-Lymphocytes/immunology , Acrylamides/pharmacology , Animals , Autophagy , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bone Marrow Transplantation/adverse effects , Coculture Techniques , Colon/abnormalities , Female , Genetic Predisposition to Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inflammatory Bowel Diseases/pathology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Necroptosis/drug effects , Nitriles , Paneth Cells/pathology , Precision Medicine , Pyrazoles/pharmacology , Pyrimidines , Radiation Chimera , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Sulfonamides/pharmacology , T-Lymphocytes/transplantation
17.
J Immunol ; 204(12): 3389-3399, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32350082

ABSTRACT

Myeloid cells are a vital component of innate immunity and comprise monocytes, macrophages, dendritic cells, and granulocytes. How myeloid cell lineage affects activation states in response to cytokines remains poorly understood. The cytokine environment and cellular infiltrate during an inflammatory response may contain prognostic features that predict disease outcome. In this study, we analyzed the transcriptional responses of human monocytes, macrophages, dendritic cells, and neutrophils in response to stimulation by IFN-γ, IFN-ß, IFN-λ, IL-4, IL-13, and IL-10 cytokines to better understand the heterogeneity of activation states in inflammatory conditions. This generated a myeloid cell-cytokine-specific response matrix that can infer representation of myeloid cells and the cytokine environment they encounter during infection, in tumors and in whole blood. Neutrophils were highly responsive to type 1 and type 2 cytokine stimulation but did not respond to IL-10. We identified transcripts specific to IFN-ß stimulation, whereas other IFN signature genes were upregulated by both IFN-γ and IFN-ß. When we used our matrix to deconvolute blood profiles from tuberculosis patients, the IFN-ß-specific neutrophil signature was reduced in tuberculosis patients with active disease, whereas the shared response to IFN-γ and IFN-ß in neutrophils was increased. When applied to glioma patients, transcripts of neutrophils exposed to IL-4/IL-13 and monocyte responses to IFN-γ or IFN-ß emerged as opposing predictors of patient survival. Hence, by dissecting how different myeloid cells respond to cytokine activation, we can delineate biological roles for myeloid cells in different cytokine environments during disease processes, especially during infection and tumor progression.


Subject(s)
Cytokines/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Tuberculosis/immunology , Cells, Cultured , Dendritic Cells/immunology , Humans , Immunity, Innate/immunology , Macrophages/immunology , Monocytes/immunology , Neoplasms/pathology , Prognosis , Tuberculosis/pathology
18.
Proc Natl Acad Sci U S A ; 116(27): 13563-13572, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31217288

ABSTRACT

The pathogen Staphylococcus aureus colonizes and infects a variety of different sites within the human body. To adapt to these different environments, S. aureus relies on a complex and finely tuned regulatory network. While some of these networks have been well-elucidated, the functions of more than 50% of the transcriptional regulators in S. aureus remain unexplored. Here, we assess the contribution of the LacI family of metabolic regulators to staphylococcal virulence. We found that inactivating the purine biosynthesis regulator purR resulted in a strain that was acutely virulent in bloodstream infection models in mice and in ex vivo models using primary human neutrophils. Remarkably, these enhanced pathogenic traits are independent of purine biosynthesis, as the purR mutant was still highly virulent in the presence of mutations that disrupt PurR's canonical role. Through the use of transcriptomics coupled with proteomics, we revealed that a number of virulence factors are differentially regulated in the absence of purR Indeed, we demonstrate that PurR directly binds to the promoters of genes encoding virulence factors and to master regulators of virulence. These results guided us into further ex vivo and in vivo studies, where we discovered that S. aureus toxins drive the death of human phagocytes and mice, whereas the surface adhesin FnbA contributes to the increased bacterial burden observed in the purR mutant. Thus, S. aureus repurposes a metabolic regulator to directly control the expression of virulence factors, and by doing so, tempers its pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Purines/biosynthesis , Repressor Proteins/metabolism , Staphylococcus aureus/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial/physiology , Humans , Mice , Repressor Proteins/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Transcription Factors/metabolism , Transcription Factors/physiology , Virulence Factors/physiology
19.
Proc Natl Acad Sci U S A ; 116(5): 1745-1754, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30635416

ABSTRACT

The past two decades have witnessed an alarming expansion of staphylococcal disease caused by community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA). The factors underlying the epidemic expansion of CA-MRSA lineages such as USA300, the predominant CA-MRSA clone in the United States, are largely unknown. Previously described virulence and antimicrobial resistance genes that promote the dissemination of CA-MRSA are carried by mobile genetic elements, including phages and plasmids. Here, we used high-resolution genomics and experimental infections to characterize the evolution of a USA300 variant plaguing a patient population at increased risk of infection to understand the mechanisms underlying the emergence of genetic elements that facilitate clonal spread of the pathogen. Genetic analyses provided conclusive evidence that fitness (manifest as emergence of a dominant clone) changed coincidently with the stepwise emergence of (i) a unique prophage and mutation of the regulator of the pyrimidine nucleotide biosynthetic operon that promoted abscess formation and colonization, respectively, thereby priming the clone for success; and (ii) a unique plasmid that conferred resistance to two topical microbiocides, mupirocin and chlorhexidine, frequently used for decolonization and infection prevention. The resistance plasmid evolved through successive incorporation of DNA elements from non-S. aureus spp. into an indigenous cryptic plasmid, suggesting a mechanism for interspecies genetic exchange that promotes antimicrobial resistance. Collectively, the data suggest that clonal spread in a vulnerable population resulted from extensive clinical intervention and intense selection pressure toward a pathogen lifestyle that involved the evolution of consequential mutations and mobile genetic elements.


Subject(s)
Community-Acquired Infections/microbiology , Methicillin-Resistant Staphylococcus aureus/genetics , Virulence/genetics , Animals , Anti-Bacterial Agents/pharmacology , Child , Chlorhexidine/pharmacology , Community-Acquired Infections/drug therapy , Genome, Bacterial/genetics , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Microbial Sensitivity Tests/methods , Mupirocin/pharmacology , Phylogeny , Plasmids/genetics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
20.
J Biol Chem ; 295(18): 5944-5959, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32179646

ABSTRACT

The rapid emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains poses a major threat to public health. MRSA possesses an arsenal of secreted host-damaging virulence factors that mediate pathogenicity and blunt immune defenses. Panton-Valentine leukocidin (PVL) and α-toxin are exotoxins that create lytic pores in the host cell membrane. They are recognized as being important for the development of invasive MRSA infections and are thus potential targets for antivirulence therapies. Here, we report the high-resolution X-ray crystal structures of both PVL and α-toxin in their soluble, monomeric, and oligomeric membrane-inserted pore states in complex with n-tetradecylphosphocholine (C14PC). The structures revealed two evolutionarily conserved phosphatidylcholine-binding mechanisms and their roles in modulating host cell attachment, oligomer assembly, and membrane perforation. Moreover, we demonstrate that the soluble C14PC compound protects primary human immune cells in vitro against cytolysis by PVL and α-toxin and hence may serve as the basis for the development of an antivirulence agent for managing MRSA infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Discovery , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Molecular Docking Simulation , Protein Domains , Protein Multimerization , Protein Structure, Quaternary , Small Molecule Libraries/metabolism , Virulence/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL