Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003310

ABSTRACT

Attributed to the tropism for host microvascular endothelium lining the blood vessels, vascular inflammation and dysfunction represent salient features of rickettsial pathogenesis, yet the details of fundamentally important pathogen interactions with host endothelial cells (ECs) as the primary targets of infection remain poorly appreciated. Mechanistic target of rapamycin (mTOR), a serine/threonine protein kinase of the phosphatidylinositol kinase-related kinase family, assembles into two functionally distinct complexes, namely mTORC1 (Raptor) and mTORC2 (Rictor), implicated in the determination of innate immune responses to intracellular pathogens via transcriptional regulation. In the present study, we investigated activation status of mTOR and its potential contributions to host EC responses during Rickettsia rickettsii and R. conorii infection. Protein lysates from infected ECs were analyzed for threonine 421/serine 424 phosphorylation of p70 S6 kinase (p70 S6K) and that of serine 2448 on mTOR itself as established markers of mTORC1 activation. For mTORC2, we assessed phosphorylation of protein kinase B (PKB or Akt) and protein kinase C (PKC), respectively, on serine 473 and serine 657. The results suggest increased phosphorylation of p70 S6K and mTOR during Rickettsia infection of ECs as early as 3 h and persisting for up to 24 h post-infection. The steady-state levels of phospho-Akt and phospho-PKC were also increased. Infection with pathogenic rickettsiae also resulted in the formation of microtubule-associated protein 1A/1B-light chain 3 (LC3-II) puncta and increased lipidation of LC3-II, a response significantly inhibited by introduction of siRNA targeting mTORC1 into ECs. These findings thus yield first evidence for the activation of both mTORC1 and mTORC2 during EC infection in vitro with Rickettsia species and suggest that early induction of autophagy in response to intracellular infection might be regulated by this important pathway known to function as a central integrator of cellular immunity and inflammation.


Subject(s)
Immunity, Innate/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Rickettsiaceae/genetics , Spotted Fever Group Rickettsiosis/genetics , Endothelial Cells/microbiology , Endothelium/metabolism , Endothelium/microbiology , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Rickettsiaceae/pathogenicity , Signal Transduction , Spotted Fever Group Rickettsiosis/microbiology , Spotted Fever Group Rickettsiosis/pathology , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
2.
J Immunol ; 204(9): 2552-2561, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32205425

ABSTRACT

The adaptive immune function of lymph nodes is dependent on constant recirculation of lymphocytes. In this article, we identify neutrophils present in the lymph node at steady state, exhibiting the same capacity for recirculation. In germ-free mice, neutrophils still recirculate through lymph nodes, and in mice cohoused with wild microbiome mice, the level of neutrophils in lymph nodes increases significantly. We found that at steady state, neutrophils enter the lymph node entirely via L-selectin and actively exit via efferent lymphatics via an S1P dependent mechanism. The small population of neutrophils in the lymph node can act as reconnaissance cells to recruit additional neutrophils in the event of bacterial dissemination to the lymph node. Without these reconnaissance cells, there is a delay in neutrophil recruitment to the lymph node and a reduction in swarm formation following Staphylococcus aureus infection. This ability to recruit additional neutrophils by lymph node neutrophils is initiated by LTB4. This study establishes the capacity of neutrophils to recirculate, much like lymphocytes via L-selectin and high endothelial venules in lymph nodes and demonstrates how the presence of neutrophils at steady state fortifies the lymph node in case of an infection disseminating through lymphatics.


Subject(s)
Lymph Nodes/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Staphylococcal Infections/immunology , Animals , Endothelium/immunology , Endothelium/microbiology , Female , L-Selectin/immunology , Lymph Nodes/microbiology , Lymphatic Vessels/immunology , Lymphatic Vessels/microbiology , Lymphocytes/immunology , Lymphocytes/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota/immunology , Sphingosine-1-Phosphate Receptors/immunology , Staphylococcal Infections/microbiology , Venules/immunology , Venules/microbiology
3.
Biofabrication ; 12(2): 025012, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31994489

ABSTRACT

Pneumonia is one of the most common infectious diseases worldwide. The influenza virus can cause severe epidemics, which results in significant morbidity and mortality. Beyond the virulence of the virus itself, epidemiological data suggest that bacterial co-infections are the major cause of increased mortality. In this context, Staphylococcus aureus represents a frequent causative bacterial pathogen. Currently available models have several limitations in the analysis of the pathogenesis of infections, e.g. some bacterial toxins strongly act in a species-specific manner. Human 2D mono-cell culture models often fail to maintain the differentiation of alveolus-specific functions. A detailed investigation of the underlying pathogenesis mechanisms requires a physiological interaction of alveolus-specific cell types. The aim of the present work was to establish a human in vitro alveolus model system composed of vascular and epithelial cell structures with cocultured macrophages resembling the human alveolus architecture and functions. We demonstrate that high barrier integrity maintained for up to 14 d in our model containing functional tissue-resident macrophages. We show that flow conditions and the presence of macrophages increased the barrier function. The infection of epithelial cells induced a high inflammatory response that spread to the endothelium. Although the integrity of the epithelium was not compromised by a single infection or co-infection, we demonstrated significant endothelial cell damage associated with loss of barrier function. We established a novel immune-responsive model that reflects the complex crosstalk between pathogens and host. The in vitro model allows for the monitoring of spatiotemporal spreading of the pathogens and the characterization of morphological and functional alterations attributed to infection. The alveolus-on-a-chip represents a promising platform for mechanistic studies of host-pathogen interactions and the identification of molecular and cellular targets of novel treatment strategies in pneumonia.


Subject(s)
Endothelium/microbiology , Endothelium/virology , Influenza, Human/virology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/virology , Staphylococcal Infections/microbiology , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Endothelium/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/virology , Humans , Influenza, Human/immunology , Lab-On-A-Chip Devices , Models, Biological , Orthomyxoviridae/physiology , Pulmonary Alveoli/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology
4.
PLoS One ; 14(12): e0226126, 2019.
Article in English | MEDLINE | ID: mdl-31815949

ABSTRACT

Salmonella can appear in the bloodstream within CD18 expressing phagocytes following oral ingestion in as little as 15 minutes. Here, we provide evidence that the process underlying this phenomenon is reverse transmigration. Reverse transmigration is a normal host process in which dendritic cells can reenter the bloodstream by traversing endothelium in the basal to apical direction. We have developed an in vitro reverse transmigration assay in which dendritic cells are given the opportunity to cross endothelial monolayers in the basal to apical direction grown on membranes with small pores, modeling how such cells can penetrate the bloodstream. We demonstrate that exposing dendritic cells to microbial components negatively regulates reverse transmigration. We propose that microbial components normally cause the host to toggle between positively and negatively regulating reverse transmigration, balancing the need to resolve inflammation with inhibiting the spread of microbes. We show that Salmonella in part overcomes this negative regulation of reverse transmigration with the Salmonella pathogenicity island-2 encoded type III secretion system, which increases reverse transmigration by over an order of magnitude. The SPI-2 type III secretion system does this in part, but not entirely by injecting the type III effector SpvC into infected cells. We further demonstrate that SpvC greatly promotes early extra-intestinal dissemination in mice. This result combined with the previous observation that the spv operon is conserved amongst strains of non-typhoidal Salmonella capable of causing bacteremia in humans suggests that this pathway to the bloodstream could be important for understanding human infections.


Subject(s)
Carbon-Oxygen Lyases/metabolism , Salmonella/metabolism , Transendothelial and Transepithelial Migration/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CD18 Antigens/deficiency , CD18 Antigens/genetics , Carbon-Oxygen Lyases/genetics , Dendritic Cells/microbiology , Dendritic Cells/physiology , Endothelium/cytology , Endothelium/microbiology , Female , Intestines/microbiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis , Phagocytes/metabolism , Phagocytes/microbiology , Salmonella/pathogenicity
5.
Clin Lab ; 65(9)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31532089

ABSTRACT

BACKGROUND: Endothelial injury is the key to the occurrence and development of bacterial infections. The aim of this study was to discuss the relationship between the molecular marker of endothelial damage, thrombomodulin (TM), and infectious disease severity, and prognosis. METHODS: From January 2017 to April 2018, 296 patients with bacterial infections and 163 controls were recruited from our hospital. The concentrations of thrombomodulin and other routine coagulation and inflammatory factors were quantified. RESULTS: Plasma levels of thrombomodulin were obviously increased in infection group compared with control group (8.30 (7.23 - 9.68) vs. 15.83 (10.60 - 22.20) TU/mL, p < 0.001) and logistic regression analysis showed that the thrombomodulin was an independent risk factor for bacterial infection (OR, 1.189 (1.079 - 1.311)). In the infection group, patients with elevated thrombomodulin levels (> 75th percentile of its distribution, n = 71) experienced a higher level of coagulation factors (p < 0.05) and inflammatory factors (p < 0.05) than patients with levels below this cutoff. Multiple linear regression analysis showed that there was a positive correlation among the plasma thrombomodulin and D-dimer, white blood cells, and procalcitonin (ß coefficient = 0.590, 0.220, and 0.208, p = 0.004, 0.027, and 0.025, respectively). With the increase of severity of disease, thrombomodulin levels gradually rose (13.58 ± 0.47 TU/mL vs. 25.07 ± 2.01 TU/mL vs. 31.34 ± 2.56 TU/mL, respectively, p < 0.001). Furthermore, there was an abnormal increase of plasma thrombomodulin in patients with bacterial infections and poor prognosis (p < 0.05). The area under curve of thrombomodulin as diagnosis for organ failure and non-survivor was 0.867 and 0.778, respectively. CONCLUSIONS: Plasmatic level of thrombomodulin could be considered as a diagnostic tool for bacterial infections. An increase in thrombomodulin plasmatic level was associated with poor outcome.


Subject(s)
Bacterial Infections/diagnosis , Biomarkers/blood , Endothelium/pathology , Thrombomodulin/blood , Aged , Bacterial Infections/blood , Bacterial Infections/microbiology , Blood Coagulation , Endothelium/microbiology , Endothelium/physiopathology , Female , Humans , Male , Middle Aged , Prognosis , Severity of Illness Index
6.
Microbiol Spectr ; 7(2)2019 03.
Article in English | MEDLINE | ID: mdl-30848239

ABSTRACT

A wide variety of pathogens reach the circulatory system during viral, parasitic, fungal, and bacterial infections, causing clinically diverse pathologies. Such systemic infections are usually severe and frequently life-threatening despite intensive care, in particular during the age of antibiotic resistance. Because of its position at the interface between the blood and the rest of the organism, the endothelium plays a central role during these infections. Using several examples of systemic infections, we explore the diversity of interactions between pathogens and the endothelium. These examples reveal that bacterial pathogens target specific vascular beds and affect most aspects of endothelial cell biology, ranging from cellular junction stability to endothelial cell proliferation and inflammation.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , Endothelium/microbiology , Animals , Bacterial Infections/blood , Drug Resistance, Bacterial , Endothelium, Vascular/microbiology , Host-Pathogen Interactions , Humans
7.
ACS Appl Mater Interfaces ; 11(10): 10337-10350, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30753784

ABSTRACT

Tunable/sustained drug loading/releasing are of significance in addressing low cytotoxicity, long-term performance, and localized mild healing response in biomedical applications. With an ingenious design, a self-healing sandwiched layer-by-layer (LBL) coating was constructed by using chitosan/heparin as adopted polyelectrolytes with embedding of micelles, in which the chitosan backbone was grafted with catechol and the micelle was modified with exposed phenylboronic acid, endowing the coating with enhanced stability by abundant interactions among coating components (e.g., boric acid ester bond formation, weak intermolecular cross-linking, π-π interactions, and H-bonding). Moreover, rapamycin and atorvastatin calcium were selected as drug candidates and loaded into micelles, followed by drug-releasing behavior study. It was found that the LBL coating maintained a linear growth mode up to 30 cycles, giving a favorable tunability of coating construction and drug loading. The coating could also support sustained release of payloads and provide wild tissue response. With the systematic in vitro and in vivo study, such catechol-phenylboronic acid-enhanced LBL coating with drug loading would also address enhanced antiplatelet adhesion/activation and direct cell fate of endothelial cells and smooth muscle cells via tuning of coating cycles and loaded drugs. With modular assembly, such coating indicated potential for achieving enhanced re-endothelialization for vascular implants.


Subject(s)
Anti-Bacterial Agents/chemistry , Chitosan/chemistry , Coated Materials, Biocompatible/chemistry , Delayed-Action Preparations/chemistry , Endothelium/drug effects , Anti-Bacterial Agents/pharmacology , Boronic Acids/chemistry , Catechols/chemistry , Chitosan/pharmacology , Coated Materials, Biocompatible/pharmacology , Delayed-Action Preparations/pharmacology , Drug Liberation , Endothelium/growth & development , Endothelium/microbiology , Humans , Micelles , Prostheses and Implants/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity
8.
Cell ; 174(1): 143-155.e16, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29779947

ABSTRACT

Neisseria meningitidis, a bacterium responsible for meningitis and septicemia, proliferates and eventually fills the lumen of blood capillaries with multicellular aggregates. The impact of this aggregation process and its specific properties are unknown. We first show that aggregative properties are necessary for efficient infection and study their underlying physical mechanisms. Micropipette aspiration and single-cell tracking unravel unique features of an atypical fluidized phase, with single-cell diffusion exceeding that of isolated cells. A quantitative description of the bacterial pair interactions combined with active matter physics-based modeling show that this behavior relies on type IV pili active dynamics that mediate alternating phases of bacteria fast mutual approach, contact, and release. These peculiar fluid properties proved necessary to adjust to the geometry of capillaries upon bacterial proliferation. Intermittent attractive forces thus generate a fluidized phase that allows for efficient colonization of the blood capillary network during infection.


Subject(s)
Bacterial Adhesion/physiology , Capillaries/microbiology , Fimbriae, Bacterial/physiology , Neisseria meningitidis/pathogenicity , Animals , Bacterial Load , Capillaries/pathology , Endothelium/metabolism , Endothelium/microbiology , Endothelium/pathology , Female , Fimbriae Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, SCID , Microscopy, Confocal , Neisseria meningitidis/physiology , Skin Transplantation , Surface Tension , Time-Lapse Imaging , Transplantation, Heterologous
9.
J Infect ; 77(1): 60-67, 2018 07.
Article in English | MEDLINE | ID: mdl-29746945

ABSTRACT

OBJECTIVES: Typhoid fever caused by Salmonella Typhi remains a major burden worldwide. Gastrointestinal bleeding can be seen in up to 10 percent of patients and may be fatal. The coagulopathy, which may be the driver of this severe complication in patients with typhoid fever, however is ill defined. The aim of this study was to evaluate the activation of coagulation, anticoagulation, and fibrinolysis in patients with acute typhoid fever. METHODS: Parameters of coagulation and fibrinolysis were measured in 28 hospitalized patients with culture-confirmed or PCR-confirmed typhoid fever and compared to 38 age- and sex-matched healthy volunteers. RESULTS: Patients demonstrated activation of the coagulation system, as reflected by elevated in vitro thrombin generation and high plasma levels of fibrinogen, D-dimer and prothrombin fragment F1 + 2 in concert with consumption of coagulation factors resulting in a prolonged prothrombin-time and activated-partial-thromboplastin-time. Concurrently, the anticoagulant proteins, protein C and antithrombin, were significantly lower in comparison to healthy controls. Patients also demonstrated evidence of activation and inhibition of fibrinolysis and a marked activation of endothelial cells. The extent of coagulation activation was associated with the course of the disease, repeated testing during convalescence showed a return toward normal values. CONCLUSIONS: Activation of coagulation is an important clinical feature of typhoid fever and is associated with severity of disease.


Subject(s)
Blood Coagulation , Endothelium/pathology , Fibrinolysis , Typhoid Fever/blood , Typhoid Fever/complications , Adult , Anticoagulants , Bangladesh , Endothelial Cells/microbiology , Endothelial Cells/pathology , Endothelium/cytology , Endothelium/microbiology , Female , Fibrin Fibrinogen Degradation Products/analysis , Humans , Male , Middle Aged , Peptide Fragments/blood , Polymerase Chain Reaction , Prospective Studies , Prothrombin , Salmonella typhi/genetics , Salmonella typhi/isolation & purification , Severity of Illness Index , Thrombocytopenia , Typhoid Fever/pathology , Young Adult
10.
Sci Rep ; 8(1): 6725, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29712976

ABSTRACT

HIV infection and type 2 diabetes are associated with altered gut microbiota, chronic inflammation, and increased cardiovascular risk. We aimed to investigate the combined effect of these diseases on gut microbiota composition and related metabolites, and a potential relation to endothelial dysfunction in individuals with HIV-infection only (n = 23), diabetes only (n = 16) or both conditions (n = 21), as well as controls (n = 24). Fecal microbiota was analyzed by Illumina sequencing of the 16 S rRNA gene. Markers of endothelial dysfunction (asymmetric dimethylarginine [ADMA]), tryptophan catabolism (kynurenine/tryptophan [KT]-ratio), and inflammation (neopterin) were measured by liquid chromatography-tandem mass spectrometry. The combination of HIV and type 2 diabetes was associated with reduced gut microbiota diversity, increased plasma KT-ratio and neopterin. Microbial genes related to tryptophan metabolism correlated with KT-ratio and low alpha diversity, in particular in HIV-infected with T2D. In multivariate analyses, KT-ratio associated with ADMA (ß = 4.58 [95% CI 2.53-6.63], p < 0.001), whereas microbiota composition per se was not associated with endothelial dysfunction. Our results indicate that tryptophan catabolism may be related to endothelial dysfunction, with a potentially detrimental interaction between HIV and diabetes. The potential contribution of gut microbiota and the impact for cardiovascular risk should be further explored in prospective studies powered for clinical end points.


Subject(s)
Cardiovascular Diseases/microbiology , Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/genetics , HIV Infections/microbiology , Inflammation/microbiology , Arginine/analogs & derivatives , Arginine/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelium/metabolism , Endothelium/microbiology , Endothelium/pathology , Feces/microbiology , Female , HIV Infections/complications , HIV Infections/genetics , HIV Infections/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Kynurenine/metabolism , Male , Metabolism/genetics , Middle Aged , Neopterin/metabolism , RNA, Ribosomal, 16S/genetics , Risk Factors , Tryptophan/metabolism
11.
PLoS One ; 12(8): e0183181, 2017.
Article in English | MEDLINE | ID: mdl-28806774

ABSTRACT

Rickettsial infections continue to cause serious morbidity and mortality in severe human cases around the world. Host cell adhesion and invasion is an essential requisite for intracellular growth, replication, and subsequent dissemination of pathogenic rickettsiae. Heparan sulfate proteoglycans [HSPGs] facilitate the interactions between fibroblast growth factor(s) and their tyrosine kinase receptors resulting in receptor dimerization/activation and have been implicated in bacterial adhesion to target host cells. In the present study, we have investigated the contributions of fibroblast growth factor receptors [FGFRs] in rickettsial entry into the host cells. Inhibition of HSPGs by heparinase and FGFRs by AZD4547 (a selective small-molecule inhibitor) results in significant reduction in rickettsial internalization into cultured human microvascular endothelial cells (ECs), which represent the primary targets of pathogenic rickettsiae during human infections. Administration of AZD4547 during R. conorii infection in a murine model of endothelial-target spotted fever rickettsiosis also diminishes pulmonary rickettsial burden in comparison to mock-treated controls. Silencing of FGFR1 expression using a small interfering RNA also leads to similar inhibition of R. rickettsii invasion into ECs. Consistent with these findings, R. rickettsii infection of ECs also results in phosphorylation of tyrosine 653/654, suggesting activation of FGFR1. Using isobaric tag for relative and absolute quantitation [iTRAQ]-based proteomics approach, we further demonstrate association of ß-peptide of rickettsial outer membrane protein OmpA with FGFR1. Mechanistically, FGFR1 binds to caveolin-1 and mediates bacterial entry via caveolin-1 dependent endocytosis. Together, these results identify host cell FGFR1 and rickettsial OmpA as another novel receptor-ligand pair contributing to the internalization of pathogenic rickettsiae into host endothelial cells and the potential application of FGFR-inhibitor drugs as adjunct therapeutics against spotted fever rickettsioses.


Subject(s)
Boutonneuse Fever/metabolism , Boutonneuse Fever/microbiology , Endocytosis , Endothelium/microbiology , Host-Pathogen Interactions , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Rickettsia/physiology , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Caveolin 1/metabolism , Caveolin 2/metabolism , Disease Models, Animal , Endothelium/pathology , Gene Knockdown Techniques , Heparan Sulfate Proteoglycans/metabolism , Humans , Peptides/chemistry , Peptides/metabolism , Protein Binding
12.
Sci Rep ; 7(1): 1538, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28484210

ABSTRACT

Treponema pallidum subsp. pallidum, the causative agent of syphilis, is a highly invasive spirochete pathogen that uses the vasculature to disseminate throughout the body. Identification of bacterial factors promoting dissemination is crucial for syphilis vaccine development. An important step in dissemination is bacterial adhesion to blood vessel surfaces, a process mediated by bacterial proteins that can withstand forces imposed on adhesive bonds by blood flow (vascular adhesins). The study of T. pallidum vascular adhesins is hindered by the uncultivable nature of this pathogen. We overcame these limitations by expressing T. pallidum adhesin Tp0751 (pallilysin) in an adhesion-attenuated strain of the cultivable spirochete Borrelia burgdorferi. Under fluid shear stress representative of conditions in postcapillary venules, Tp0751 restored bacterial-vascular interactions to levels similar to those observed for infectious B. burgdorferi and a gain-of-function strain expressing B. burgdorferi vascular adhesin BBK32. The strength and stability of Tp0751- and BBK32-dependent endothelial interactions under physiological shear stress were similar, although the mechanisms stabilizing these interactions were distinct. Tp0751 expression also permitted bacteria to interact with postcapillary venules in live mice as effectively as BBK32-expressing strains. These results demonstrate that Tp0751 can function as a vascular adhesin.


Subject(s)
Adhesins, Bacterial/metabolism , Borrelia burgdorferi/genetics , Gene Expression , Lyme Disease/microbiology , Treponema pallidum/metabolism , Venules/microbiology , Animals , Bacterial Adhesion , Endothelium/microbiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Shear Strength , Stress, Mechanical
13.
Microb Pathog ; 105: 326-333, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28286149

ABSTRACT

Francisella noatunensis subsp. orientalis (Fno) (syn. F. asiatica) is an emergent warmwater fish pathogen and the causative agent of piscine francisellosis. Although Fno causes septicemia and can live extracellularly in infected tilapia (Oreochromis spp.), the early interaction of Fno with vasculature endothelium is unknown. In the present study, we examined the interaction of wild-type Fno (WT) and two Fno knockout [intracellular growth loci C (ΔiglC) and pathogenicity determinant protein A (ΔpdpA)] strains with the endothelial O. mossambicus bulbus arteriosus cell line (TmB) at 25 °C and 30 °C. Similar amounts of WT, ΔiglC, and ΔpdpA attached and were detected intracellularly after 5 h of incubation at both temperatures; however temperature affected attachment and uptake. While significantly greater amounts of Fno (WT, ΔiglC, and ΔpdpA) were detected intracellularly when TmB cells were incubated at 30 °C, bacteria attached to TmBs at greater levels at 25 °C. Only WT Fno was able to replicate intracellularly at 25 °C, which resulted in Fno mediated cytotoxicity and apoptosis at 24 and 72 h post-infection. WT Fno incubated at 30 °C as well as ΔiglC, and ΔpdpA incubated at 25 °C and 30 °C were all defective for survival, replication, and the ability to cause cytotoxicity in TmB. Taken together, these results demonstrate that temperature plays a vital role for Fno intracellular survival, persistence and cytotoxicity.


Subject(s)
Fish Diseases/microbiology , Francisella/physiology , Tilapia/microbiology , Adhesins, Bacterial/genetics , Animals , Bacterial Proteins/genetics , Cell Line , Endothelium/microbiology , Fish Diseases/pathology , Francisella/genetics , Francisella/growth & development , Gene Knockout Techniques , Genome, Bacterial , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Gram-Negative Bacterial Infections/veterinary , Host-Pathogen Interactions , Mutation
14.
mBio ; 5(3): e01101-14, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24895304

ABSTRACT

UNLABELLED: Cryptococcus spp. cause life-threatening fungal infection of the central nervous system (CNS), predominantly in patients with a compromised immune system. Why Cryptococcus neoformans has this remarkable tropism for the CNS is not clear. Recent research on cerebral pathogenesis of C. neoformans revealed a predominantly transcellular migration of cryptococci across the brain endothelium; however, the identities of key fungal virulence factors that function specifically to invade the CNS remain unresolved. Here we found that a novel, secreted metalloprotease (Mpr1) that we identified in the extracellular proteome of C. neoformans (CnMpr1) is required for establishing fungal disease in the CNS. Mpr1 belongs to a poorly characterized M36 class of fungalysins that are expressed in only some fungal species. A strain of C. neoformans lacking the gene encoding Mpr1 (mpr1Δ) failed to breach the endothelium in an in vitro model of the human blood-brain barrier (BBB). A mammalian host infected with the mpr1Δ null strain demonstrated significant improvement in survival due to a reduced brain fungal burden and lacked the brain pathology commonly associated with cryptococcal disease. The in vivo studies further indicate that Mpr1 is not required for fungal dissemination and Mpr1 likely targets the brain endothelium specifically. Remarkably, the sole expression of CnMPR1 in Saccharomyces cerevisiae resulted in a robust migration of yeast cells across the brain endothelium, demonstrating Mpr1's specific activity in breaching the BBB and suggesting that Mpr1 may function independently of the hyaluronic acid-CD44 pathway. This distinct role for Mpr1 may develop into innovative treatment options and facilitate a brain-specific drug delivery platform. IMPORTANCE: Cryptococcus neoformans is a medically relevant fungal pathogen causing significant morbidity and mortality, particularly in immunocompromised individuals. An intriguing feature is its strong neurotropism, and consequently the hallmark of cryptococcal disease is a brain infection, cryptococcal meningoencephalitis. For C. neoformans to penetrate the central nervous system (CNS), it first breaches the blood-brain barrier via a transcellular pathway; however, the identities of fungal factors required for this transmigration remain largely unknown. In an effort to identify extracellular fungal proteins that could mediate interactions with the brain endothelium, we undertook a proteomic analysis of the extracellular proteome and identified a secreted metalloprotease (Mpr1) belonging to the M36 class of fungalysins. Here we found that Mpr1 promotes migration of C. neoformans across the brain endothelium and into the CNS by facilitating attachment of cryptococci to the endothelium surface, thus underscoring the critical role of M36 proteases in fungal pathogenesis.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/metabolism , Meningoencephalitis/microbiology , Metalloproteases/metabolism , Animals , Blood-Brain Barrier/microbiology , Brain/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/ultrastructure , Disease Models, Animal , Endothelium/microbiology , Extracellular Space/metabolism , Fungal Proteins/genetics , Gene Expression , Meningoencephalitis/pathology , Metalloproteases/genetics , Mice , Virulence Factors/metabolism
15.
Immunity ; 40(4): 463-75, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24745331

ABSTRACT

Sepsis, a clinical syndrome occurring in patients following infection or injury, is a leading cause of morbidity and mortality worldwide. Current immunological mechanisms do not explain the basis of cellular dysfunction and organ failure, the ultimate cause of death. Here we review current dogma and argue that it is time to delineate novel immunometabolic and neurophysiological mechanisms underlying the altered cellular bioenergetics and failure of epithelial and endothelial barriers that produce organ dysfunction and death. These mechanisms might hold the key to future therapeutic strategies.


Subject(s)
Endothelium/immunology , Epithelium/immunology , Sepsis/immunology , Animals , Endothelium/microbiology , Energy Metabolism , Epithelium/microbiology , Humans , Inflammation Mediators/metabolism , Multiple Organ Failure , Neuroimmunomodulation , Synaptic Transmission/immunology
16.
Thromb Haemost ; 111(5): 970-80, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24401906

ABSTRACT

Pneumococcal pneumonia is a frequent cause of gram-positive sepsis and has a high mortality. The endothelial protein C receptor (EPCR) has been implicated in both the activation of protein C (PC) and the anti-inflammatory actions of activated (A)PC. The aim of this study was to determine the role of the EPCR in murine pneumococcal pneumonia and sepsis. Wild-type (WT), EPCR knockout (KO) and Tie2-EPCR mice, which overexpress EPCR on the endothelium, were infected intranasally (pneumonia) or intravenously (sepsis) with viable Streptococcus pneumoniae and euthanised at 24 or 48 hours after initiation of the infection for analyses. Pneumonia did not alter constitutive EPCR expression on pulmonary endothelium but was associated with an influx of EPCR positive neutrophils into lung tissue. In pneumococcal pneumonia EPCR KO mice demonstrated diminished bacterial growth in the lungs and dissemination to spleen and liver, reduced neutrophil recruitment to the lungs and a mitigated inflammatory response. Moreover, EPCR KO mice displayed enhanced activation of coagulation in the early phase of disease. Correspondingly, in pneumococcal sepsis EPCR KO mice showed reduced bacterial growth in lung and liver and attenuated cytokine release. Conversely, EPCR-overexpressing mice displayed higher bacterial outgrowth in lung, blood, spleen and liver in pneumococcal sepsis. In conclusion, EPCR impairs antibacterial defense in both pneumococcal pneumonia and sepsis, which is associated with an enhanced pro-inflammatory response.


Subject(s)
Endothelium/metabolism , Lung/pathology , Neutrophils/metabolism , Pneumonia, Pneumococcal/immunology , Receptors, Cell Surface/metabolism , Sepsis/immunology , Streptococcus pneumoniae/immunology , Animals , Bacterial Load/genetics , Cell Movement/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Protein C Receptor , Endothelium/immunology , Endothelium/microbiology , Female , Humans , Immunity, Innate/genetics , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Neutrophils/immunology , Neutrophils/microbiology , Pneumonia, Pneumococcal/microbiology , Receptors, Cell Surface/genetics , Streptococcus pneumoniae/growth & development
17.
Cell Microbiol ; 15(4): 512-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23189983

ABSTRACT

The brain and meningeal spaces are protected from bacterial invasion by the blood-brain barrier, formed by specialized endothelial cells and tight intercellular junctional complexes. However, once in the bloodstream, Neisseria meningitidis crosses this barrier in about 60% of the cases. This highlights the particular efficacy with which N. meningitidis targets the brain vascular cell wall. The first step of central nervous system invasion is the direct interaction between bacteria and endothelial cells. This step is mediated by the type IV pili, which induce a remodelling of the endothelial monolayer, leading to the opening of the intercellular space. In this review, strategies used by the bacteria to survive in the bloodstream, to colonize the brain vasculature and to cross the blood-brain barrier will be discussed.


Subject(s)
Blood-Brain Barrier/microbiology , Brain/microbiology , Cerebrospinal Fluid/microbiology , Endothelium/microbiology , Neisseria meningitidis/physiology , Blood-Brain Barrier/immunology , Fimbriae, Bacterial/metabolism , Host-Pathogen Interactions , Neisseria meningitidis/growth & development
18.
PLoS Pathog ; 8(10): e1002947, 2012.
Article in English | MEDLINE | ID: mdl-23055927

ABSTRACT

The serine-rich repeat glycoprotein Srr1 of Streptococcus agalactiae (GBS) is thought to be an important adhesin for the pathogenesis of meningitis. Although expression of Srr1 is associated with increased binding to human brain microvascular endothelial cells (hBMEC), the molecular basis for this interaction is not well defined. We now demonstrate that Srr1 contributes to GBS attachment to hBMEC via the direct interaction of its binding region (BR) with human fibrinogen. When assessed by Far Western blotting, Srr1 was the only protein in GBS extracts that bound fibrinogen. Studies using recombinant Srr1-BR and purified fibrinogen in vitro confirmed a direct protein-protein interaction. Srr1-BR binding was localized to amino acids 283-410 of the fibrinogen Aα chain. Structural predictions indicated that the conformation of Srr1-BR is likely to resemble that of SdrG and other related staphylococcal proteins that bind to fibrinogen through a "dock, lock, and latch" mechanism (DLL). Deletion of the predicted latch domain of Srr1-BR abolished the interaction of the BR with fibrinogen. In addition, a mutant GBS strain lacking the latch domain exhibited reduced binding to hBMEC, and was significantly attenuated in an in vivo model of meningitis. These results indicate that Srr1 can bind fibrinogen directly likely through a DLL mechanism, which has not been described for other streptococcal adhesins. This interaction was important for the pathogenesis of GBS central nervous system invasion and subsequent disease progression.


Subject(s)
Bacterial Proteins/metabolism , Brain/metabolism , Endothelium/metabolism , Fibrinogen/metabolism , Glycoproteins/metabolism , Meningitis, Bacterial/microbiology , Streptococcal Infections/microbiology , Streptococcus agalactiae/metabolism , Streptococcus agalactiae/pathogenicity , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion , Binding Sites , Brain/microbiology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelium/microbiology , Humans , Meningitis, Bacterial/metabolism , Mice , Protein Binding , Protein Conformation , Sequence Analysis, Protein , Streptococcal Infections/metabolism
19.
PLoS One ; 7(8): e42624, 2012.
Article in English | MEDLINE | ID: mdl-22952600

ABSTRACT

BACKGROUND: Circulating levels of microbial products are increased in HIV infection, and provoke endothelial dysfunction in other disease settings. METHODOLOGY/PRINCIPAL FINDINGS: We examined data from a cross-sectional single site study at Indiana University (Indiana, N = 85) and a 24- week multicenter prospective study of antiretroviral therapy (ART) initiation (ACTG 5152s, N = 75). Brachial artery flow-mediated dilation (FMD) was measured by ultrasound. Plasma lipopolysaccharide (LPS) and soluble CD14 (sCD14) levels were measured from stored specimens and correlated with FMD values using Pearson correlations. The Indiana subjects were 63% male with a mean age of 39 years and a median CD4 count of 406 cells/mm(3) (388 not on ART, 464 on ART). The 5152s subjects were 92% were male with a mean age of 35 years and a median CD4 count of 251 cells/mm(3) at entry which increased to 396 cells/mm(3) on ART. When analyzing the two cohorts individually or in combination neither sCD14 nor LPS correlated significantly with FMD. In a pre-specified subgroup analysis of the Indiana subjects receiving ART (N = 46, mean ART duration 40 months) LPS was inversely correlated with FMD (r = -0.33, p = 0.02), but not sCD14 (r = -0.01, p = 0.9). Multivariate analysis confirmed LPS as an independent predictor of FMD in this subgroup (p = 0.02). CONCLUSIONS/SIGNIFICANCE: In HIV-infected individuals on prolonged ART, higher LPS levels are associated with worse endothelial function but not in untreated subjects or at 24 weeks after ART initiation. Persistent microbial translocation may contribute to arterial dysfunction and the increased cardiovascular disease risk observed in individuals on long-term ART.


Subject(s)
Endothelium/physiopathology , HIV Infections/complications , HIV Infections/physiopathology , Adult , Anti-Retroviral Agents/therapeutic use , Biological Transport , Brachial Artery/physiopathology , Cardiovascular Diseases/complications , Cardiovascular Diseases/etiology , Cohort Studies , Cross-Sectional Studies , Endothelium/microbiology , Endothelium/virology , Female , HIV Infections/microbiology , Humans , Indiana , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/blood , Lipopolysaccharides/metabolism , Male , Multivariate Analysis , Prospective Studies , Ultrasonography/methods
20.
Cell Microbiol ; 14(8): 1166-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22519749

ABSTRACT

The Gram-negative genus Bartonella comprises arthropod-borne pathogens that typically infect mammals in a host-specific manner. Bartonella bacilliformis and Bartonella quintana are human-specific pathogens, while several zoonotic bartonellae specific for diverse animal hosts infect humans as an incidental host. Clinical manifestations of Bartonella infections range from mild symptoms to life-threatening disease. Following transmission by blood-sucking arthropods or traumatic contact with infected animals, bartonellae display sequential tropisms towards endothelial and possibly other nucleated cells and erythrocytes, the latter in a host-specific manner. Attachment to the extracellular matrix (ECM) and to nucleated cells is mediated by surface-exposed bacterial adhesins, in particular trimeric autotransporter adhesins (TAAs). The subsequent engulfment of the pathogen into a vacuolar structure follows a unique series of events whereby the pathogen avoids the endolysosomal compartments. For Bartonella henselae and assumingly most other species, the infection process is aided at different steps by Bartonella effector proteins (Beps). They are injected into host cells through the type IV secretion system (T4SS) VirB/D4 and subvert host cellular functions to favour pathogen uptake. Bacterial binding to erythrocytes is mediated by Trw, another T4SS, in a strictly host-specific manner, followed by pathogen-forced uptake involving the IalB invasin and subsequent replication and persistence within a membrane-bound intra-erythrocytic compartment.


Subject(s)
Bartonella Infections/microbiology , Bartonella/physiology , Animals , Arthropod Vectors/microbiology , Bacterial Adhesion , Endothelium/microbiology , Erythrocytes/microbiology , Host-Pathogen Interactions , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...