Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Chem Biol ; 18(3): 583-594, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36795767

ABSTRACT

Biomolecular condensates formed by liquid-liquid phase separation have been implicated in multiple diseases. Modulation of condensate dynamics by small molecules has therapeutic potential, but so far, few condensate modulators have been disclosed. The SARS-CoV-2 nucleocapsid (N) protein forms phase-separated condensates that are hypothesized to play critical roles in viral replication, transcription, and packaging, suggesting that N condensation modulators might have anti-coronavirus activity across multiple strains and species. Here, we show that N proteins from all seven human coronaviruses (HCoVs) vary in their tendency to undergo phase separation when expressed in human lung epithelial cells. We developed a cell-based high-content screening platform and identified small molecules that both promote and inhibit condensation of SARS-CoV-2 N. Interestingly, these host-targeted small molecules exhibited condensate-modulatory effects across all HCoV Ns. Some have also been reported to exhibit antiviral activity against SARS-CoV-2, HCoV-OC43, and HCoV-229E viral infections in cell culture. Our work reveals that the assembly dynamics of N condensates can be regulated by small molecules with therapeutic potential. Our approach allows for screening based on viral genome sequences alone and might enable rapid paths to drug discovery with value for confronting future pandemics.


Subject(s)
COVID-19 , Coronavirus 229E, Human , Coronavirus OC43, Human , Humans , SARS-CoV-2 , Nucleocapsid Proteins
2.
Toxins (Basel) ; 14(2)2022 02 18.
Article in English | MEDLINE | ID: mdl-35202178

ABSTRACT

The Gram-negative, opportunistic pathogen Pseudomonas aeruginosa utilizes a type III secretion system to inject exoenzyme effectors into a target host cell. Of the four best-studied exoenzymes, ExoU causes rapid cell damage and death. ExoU is a phospholipase A2 (PLA2) that hydrolyses host cell membranes, and P. aeruginosa strains expressing ExoU are associated with poor outcomes in critically ill patients with pneumonia. While the effects of ExoU on lung epithelial and immune cells are well studied, a role for ExoU in disrupting lung endothelial cell function has only recently emerged. Lung endothelial cells maintain a barrier to fluid and protein flux into tissue and airspaces and regulate inflammation. Herein, we describe a pulmonary microvascular endothelial cell (PMVEC) culture infection model to examine the effects of ExoU. Using characterized P. aeruginosa strains and primary clinical isolates, we show that strains expressing ExoU disrupt PMVEC barrier function by causing substantial PMVEC damage and lysis, in a PLA2-dependent manner. In addition, we show that strains expressing ExoU activate the pro-inflammatory caspase-1, in a PLA2-dependent manner. Considering the important roles for mitochondria and oxidative stress in regulating inflammatory responses, we next examined the effects of ExoU on reactive oxygen species production. Infection of PMVECs with P. aeruginosa strains expressing ExoU triggered a robust oxidative stress compared to strains expressing other exoenzyme effectors. We also provide evidence that, intriguingly, ExoU PLA2 activity was detectable in mitochondria and mitochondria-associated membrane fractions isolated from P. aeruginosa-infected PMVECs. Interestingly, ExoU-mediated activation of caspase-1 was partially inhibited by reactive oxygen species scavengers. Together, these data suggest ExoU exerts pleiotropic effects on PMVEC function during P. aeruginosa infection that may inhibit endothelial barrier and inflammatory functions.


Subject(s)
Bacterial Proteins/toxicity , Caspase 1/drug effects , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/physiopathology , Endothelial Cells/drug effects , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa/genetics , Caspase 1/metabolism , Genetic Variation , Genotype , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Pseudomonas Infections/genetics
3.
Infect Immun ; 90(3): e0047021, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35130452

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen that causes nosocomial pneumonia, urinary tract infections, and bacteremia. A hallmark of P. aeruginosa pathogenesis is disruption of host cell function by the type III secretion system (T3SS) and its cognate exoenzyme effectors. The T3SS effector ExoU is phospholipase A2 (PLA2) that targets the host cell plasmalemmal membrane to induce cytolysis and is an important virulence factor that mediates immune avoidance. In addition, ExoU has been shown to subvert the host inflammatory response in a noncytolytic manner. In primary bone marrow-derived macrophages (BMDMs), P. aeruginosa infection is sensed by the nucleotide-binding domain containing leucine-rich repeats-like receptor 4 (NLRC4) inflammasome, which triggers caspase-1 activation and inflammation. ExoU transiently inhibits NLRC4 inflammasome-mediated activation of caspase-1 and its downstream target, interleukin 1ß (IL-1ß), to suppress activation of inflammation. In the present study, we sought to identify additional noncytolytic virulence functions for ExoU and discovered an unexpected association between ExoU, host mitochondria, and NLRC4. We show that infection of BMDMs with P. aeruginosa strains expressing ExoU elicited mitochondrial oxidative stress. In addition, mitochondria and mitochondrion-associated membrane fractions enriched from infected cells exhibited evidence of autophagy activation, indicative of damage. The observation that ExoU elicited mitochondrial stress and damage suggested that ExoU may also associate with mitochondria during infection. Indeed, ExoU phospholipase A2 enzymatic activity was present in enriched mitochondria and mitochondrion-associated membrane fractions isolated from P. aeruginosa-infected BMDMs. Intriguingly, enriched mitochondria and mitochondrion-associated membrane fractions isolated from infected Nlrc4 homozygous knockout BMDMs displayed significantly lower levels of ExoU enzyme activity, suggesting that NLRC4 plays a role in the ExoU-mitochondrion association. These observations prompted us to assay enriched mitochondria and mitochondrion-associated membrane fractions for NLRC4, caspase-1, and IL-1ß. NLRC4 and pro-caspase-1 were detected in enriched mitochondria and mitochondrion-associated membrane fractions isolated from noninfected BMDMs, and active caspase-1 and active IL-1ß were detected in response to P. aeruginosa infection. Interestingly, ExoU inhibited mitochondrion-associated caspase-1 and IL-1ß activation. The implications of ExoU-mediated effects on mitochondria and the NLRC4 inflammasome during P. aeruginosa infection are discussed.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Caspase 1/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice , Phospholipases/metabolism , Pseudomonas aeruginosa/physiology , Type III Secretion Systems/metabolism
4.
Toxins (Basel) ; 13(12)2021 12 09.
Article in English | MEDLINE | ID: mdl-34941717

ABSTRACT

Pseudomonas aeruginosa is an opportunistic, Gram-negative pathogen and an important cause of hospital acquired infections, especially in immunocompromised patients. Highly virulent P. aeruginosa strains use a type III secretion system (T3SS) to inject exoenzyme effectors directly into the cytoplasm of a target host cell. P. aeruginosa strains that express the T3SS effector, ExoU, associate with adverse outcomes in critically ill patients with pneumonia, owing to the ability of ExoU to rapidly damage host cell membranes and subvert the innate immune response to infection. Herein, we review the structure, function, regulation, and virulence characteristics of the T3SS effector ExoU, a highly cytotoxic phospholipase A2 enzyme.


Subject(s)
Bacterial Infections/immunology , Bacterial Proteins/immunology , Host-Parasite Interactions/immunology , Immunity, Innate/drug effects , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/pathogenicity , Type III Secretion Systems/immunology , Humans
5.
Toxins (Basel) ; 12(6)2020 06 04.
Article in English | MEDLINE | ID: mdl-32512716

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that causes pneumonia in immunocompromised and intensive care unit (ICU) patients. During host infection, P. aeruginosa upregulates the type III secretion system (T3SS), which is used to intoxicate host cells with exoenzyme (Exo) virulence factors. Of the four known Exo virulence factors (U, S, T and Y), ExoU has been shown in prior studies to associate with high mortality rates. Preclinical studies have shown that ExoY is an important edema factor in lung infection caused by P. aeruginosa, although its importance in clinical isolates of P. aeruginosa is unknown. We hypothesized that expression of ExoY would be highly prevalent in clinical isolates and would significantly contribute to patient morbidity secondary to P. aeruginosa pneumonia. A single-center, prospective observational study was conducted at the University of Alabama at Birmingham Hospital. Mechanically ventilated ICU patients with a bronchoalveolar lavage fluid culture positive for P. aeruginosa were included. Enrolled patients were followed from ICU admission to discharge and clinical P. aeruginosa isolates were genotyped for the presence of exoenzyme genes. Ninety-nine patients were enrolled in the study. ExoY was present in 93% of P. aeruginosa clinical isolates. Moreover, ExoY alone (ExoY+/ExoU-) was present in 75% of P. aeruginosa isolates, compared to 2% ExoU alone (ExoY-/ExoU+). We found that bacteria isolated from human samples expressed active ExoY and ExoU, and the presence of ExoY in clinical isolates was associated with end-organ dysfunction. This is the first study we are aware of that demonstrates that ExoY is important in clinical outcomes secondary to nosocomial pneumonia.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cross Infection/microbiology , Glucosyltransferases/metabolism , Multiple Organ Failure/microbiology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/enzymology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Cells, Cultured , Critical Illness , Cross Infection/diagnosis , Cross Infection/mortality , Female , Glucosyltransferases/genetics , Humans , Male , Middle Aged , Multiple Organ Failure/diagnosis , Multiple Organ Failure/mortality , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/mortality , Prospective Studies , Pseudomonas Infections/diagnosis , Pseudomonas Infections/mortality , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Rats , Respiration, Artificial/adverse effects , Risk Factors , Virulence , Virulence Factors/genetics
6.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L1074-L1083, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186399

ABSTRACT

Activation of the inflammasome-caspase-1 axis in lung endothelial cells is emerging as a novel arm of the innate immune response to pneumonia and sepsis caused by Pseudomonas aeruginosa. Increased levels of circulating autacoids are hallmarks of pneumonia and sepsis and induce physiological responses via cAMP signaling in targeted cells. However, it is unknown whether cAMP affects other functions, such as P. aeruginosa-induced caspase-1 activation. Herein, we describe the effects of cAMP signaling on caspase-1 activation using a single cell flow cytometry-based assay. P. aeruginosa infection of cultured lung endothelial cells caused caspase-1 activation in a distinct population of cells. Unexpectedly, pharmacological cAMP elevation increased the total number of lung endothelial cells with activated caspase-1. Interestingly, addition of cAMP agonists augmented P. aeruginosa infection of lung endothelial cells as a partial explanation underlying cAMP priming of caspase-1 activation. The cAMP effect(s) appeared to function as a priming signal because addition of cAMP agonists was required either before or early during the onset of infection. However, absolute cAMP levels measured by ELISA were not predictive of cAMP-priming effects. Importantly, inhibition of de novo cAMP synthesis decreased the number of lung endothelial cells with activated caspase-1 during infection. Collectively, our data suggest that lung endothelial cells rely on cAMP signaling to prime caspase-1 activation during P. aeruginosa infection.


Subject(s)
Caspase 1/genetics , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Pseudomonas aeruginosa/metabolism , Signal Transduction , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Caspase 1/metabolism , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP/agonists , Cyclic AMP/antagonists & inhibitors , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dinoprostone/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Endothelial Cells/pathology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Primary Cell Culture , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Rats , Rolipram/pharmacology , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...