Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(4): e1012156, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38598560

ABSTRACT

SARS-CoV-2 has been shown to cause wide-ranging ocular abnormalities and vision impairment in COVID-19 patients. However, there is limited understanding of SARS-CoV-2 in ocular transmission, tropism, and associated pathologies. The presence of viral RNA in corneal/conjunctival tissue and tears, along with the evidence of viral entry receptors on the ocular surface, has led to speculation that the eye may serve as a potential route of SARS-CoV-2 transmission. Here, we investigated the interaction of SARS-CoV-2 with cells lining the blood-retinal barrier (BRB) and the role of the eye in its transmission and tropism. The results from our study suggest that SARS-CoV-2 ocular exposure does not cause lung infection and moribund illness in K18-hACE2 mice despite the extended presence of viral remnants in various ocular tissues. In contrast, intranasal exposure not only resulted in SARS-CoV-2 spike (S) protein presence in different ocular tissues but also induces a hyperinflammatory immune response in the retina. Additionally, the long-term exposure to viral S-protein caused microaneurysm, retinal pigmented epithelium (RPE) mottling, retinal atrophy, and vein occlusion in mouse eyes. Notably, cells lining the BRB, the outer barrier, RPE, and the inner barrier, retinal vascular endothelium, were highly permissive to SARS-CoV-2 replication. Unexpectedly, primary human corneal epithelial cells were comparatively resistant to SARS-CoV-2 infection. The cells lining the BRB showed induced expression of viral entry receptors and increased susceptibility towards SARS-CoV-2-induced cell death. Furthermore, hyperglycemic conditions enhanced the viral entry receptor expression, infectivity, and susceptibility of SARS-CoV-2-induced cell death in the BRB cells, confirming the reported heightened pathological manifestations in comorbid populations. Collectively, our study provides the first evidence of SARS-CoV-2 ocular tropism via cells lining the BRB and that the virus can infect the retina via systemic permeation and induce retinal inflammation.


Subject(s)
Blood-Retinal Barrier , COVID-19 , Retina , SARS-CoV-2 , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Animals , Blood-Retinal Barrier/virology , COVID-19/immunology , COVID-19/virology , Mice , Humans , Retina/virology , Retina/immunology , Retina/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology , Inflammation/immunology , Inflammation/virology , Betacoronavirus/physiology , Viral Tropism , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/pathology
2.
Infect Immun ; 90(8): e0016522, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35900096

ABSTRACT

A newly attenuated Yersinia pseudotuberculosis strain (designated Yptb1) with triple mutation Δasd ΔyopK ΔyopJ and chromosomal insertion of the Y. pestis caf1R-caf1M-caf1A-caf1 operon was constructed as a live vaccine platform. Yptb1 tailored with an Asd+ plasmid (pYA5199) (designated Yptb1[pYA5199]) simultaneously delivers Y. pestis LcrV and F1. The attenuated Yptb1(pYA5199) localized in the Peyer's patches, lung, spleen, and liver for a few weeks after oral immunization without causing any disease symptoms in immunized rodents. An oral prime-boost Yptb1(pYA5199) immunization stimulated potent antibody responses to LcrV, F1, and Y. pestis whole-cell lysate (YPL) in Swiss Webster mice and Brown Norway rats. The prime-boost Yptb1(pYA5199) immunization induced higher antigen-specific humoral and cellular immune responses in mice than a single immunization did, and it provided complete short-term and long-term protection against a high dose of intranasal Y. pestis challenge in mice. Moreover, the prime-boost immunization afforded substantial protection for Brown Norway rats against an aerosolized Y. pestis challenge. Our study highlights that Yptb1(pYA5199) has high potential as an oral vaccine candidate against pneumonic plague.


Subject(s)
Plague Vaccine , Plague , Yersinia pestis , Yersinia pseudotuberculosis Infections , Yersinia pseudotuberculosis , Animals , Antibodies, Bacterial , Antigens, Bacterial/genetics , Mice , Plague/prevention & control , Rats , Vaccination , Yersinia pestis/genetics , Yersinia pseudotuberculosis/genetics
3.
FASEB J ; 36(3): e22197, 2022 03.
Article in English | MEDLINE | ID: mdl-35147989

ABSTRACT

Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.


Subject(s)
Escherichia coli Proteins/toxicity , Hemolysin Proteins/toxicity , Inflammasomes/metabolism , Meningitis, Escherichia coli/metabolism , Neonatal Sepsis/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Cells, Cultured , Escherichia coli K12 , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Macrophages/metabolism , Meningitis, Escherichia coli/microbiology , Mice , Mice, Inbred C57BL , Neonatal Sepsis/microbiology , Receptors, Purinergic P2X/genetics
4.
Infect Immun ; 89(3)2021 02 16.
Article in English | MEDLINE | ID: mdl-33257532

ABSTRACT

Pneumonic plague, caused by Yersinia pestis, is a rapidly progressing bronchopneumonia involving focal bacterial growth, neutrophilic congestion, and alveolar necrosis. Within a short time after inhalation of Y. pestis, inflammatory cytokines are expressed via the Toll/interleukin-1 (IL-1) adaptor myeloid differentiation primary response 88 (MyD88), which facilitates the primary lung infection. We previously showed that Y. pestis lacking the 102-kb chromosomal pigmentation locus (pgm) is unable to cause inflammatory damage in the lungs, whereas the wild-type (WT) strain induces the toxic MyD88 pulmonary inflammatory response. In this work, we investigated the involvement of the pgm in skewing the inflammatory response during pneumonic plague. We show that the early MyD88-dependent and -independent cytokine responses to pgm- Y. pestis infection of the lungs are similar yet distinct from those that occur during pgm+ infection. Furthermore, we found that MyD88 was necessary to prevent growth of the iron-starved pgm- Y. pestis despite the presence of iron chelators lactoferrin and transferrin. However, while this induced neutrophil recruitment, there was no hyperinflammatory response, and pulmonary disease was mild without MyD88. In contrast, growth in blood and tissues progressed rapidly in the absence of MyD88, due to an almost total loss of serum interferon gamma (IFN-γ). We further show that the expression of MyD88 by myeloid cells is important to control bacteremia but not the primary lung infection. The combined data indicate distinct roles for myeloid and nonmyeloid MyD88 and suggest that expression of the pgm is necessary to skew the inflammatory response in the lungs to cause pneumonic plague.


Subject(s)
Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pigmentation/genetics , Pigmentation/physiology , Plague/genetics , Plague/metabolism , Yersinia pestis/genetics , Yersinia pestis/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation, Bacterial , Humans , Plague/microbiology
5.
Article in English | MEDLINE | ID: mdl-32015027

ABSTRACT

Pneumonic plague, caused by the Gram-negative bacteria Yersinia pestis, is an invasive, rapidly progressing disease with poor survival rates. Following inhalation of Y. pestis, bacterial invasion of the lungs and a tissue-damaging inflammatory response allows vascular spread of the infection. Consequently, primary pneumonic plague is a multiorgan disease involving sepsis and necrosis of immune tissues and the liver, as well as bronchopneumonia and rampant bacterial growth. Given the likely role of the hyperinflammatory response in accelerating the destruction of tissue, in this work we evaluated the therapeutic potential of the inducible cytoprotective enzyme heme oxygenase 1 (HO-1) against primary pneumonic plague. On its own, the HO-1 inducer cobalt protoporphyrin IX (CoPP) provided mice protection from lethal challenge with Y. pestis CO92 with improved pulmonary bacterial clearance and a dampened inflammatory response compared to vehicle-treated mice. Furthermore, CoPP treatment combined with doxycycline strongly enhanced protection in a rat aerosol challenge model. Compared to doxycycline alone, CoPP treatment increased survival, with a 3-log decrease in median bacterial titer recovered from the lungs and the general absence of a systemic hyperinflammatory response. In contrast, treatment with the HO-1 inhibitor SnPP had no detectable impact on doxycycline efficacy. The combined data indicate that countering inflammatory toxicity by therapeutically inducing HO-1 is effective in reducing the rampant growth of Y. pestis and preventing pneumonic plague.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Doxycycline/therapeutic use , Heme Oxygenase-1/metabolism , Plague/prevention & control , Protoporphyrins/therapeutic use , Yersinia pestis/drug effects , Aerosols , Animals , Bronchopneumonia/microbiology , Bronchopneumonia/pathology , Disease Models, Animal , Drug Therapy, Combination , Female , Heme Oxygenase-1/genetics , Humans , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Plague/drug therapy , Plague/microbiology , Rats , Rats, Sprague-Dawley , Yersinia pestis/growth & development
6.
Methods Mol Biol ; 2010: 29-39, 2019.
Article in English | MEDLINE | ID: mdl-31177429

ABSTRACT

Primary pneumonic plague occurs when Yersinia pestis is inhaled into the lower respiratory tract where it invades the alveoli and grows. Rapid bacterial growth eventually elicits a neutrophilic inflammatory response that is ineffective and damaging, leading to accelerated progression of disease. In the laboratory, modeling of primary pneumonic plague can be accomplished by instillation of bacterial culture in the nares of anesthetized mice and rats. Although primary pneumonic plague can develop from this method, variability in dosing and side effects of anesthesia can complicate data interpretation. In contrast, aerosol challenge models allow for well-controlled studies of pneumonic plague with minimal experimental bias and unwanted side effects. For these reasons, antibiotic testing and the licensing of new treatments depend on efficacy data generated from aerosol delivery of Y. pestis in order to more accurately model transmission and the early stages of human pneumonic plague. In order to meet this need, we have extensively characterized pneumonic plague in mice and rats challenged by nose-only exposure to Yersinia pestis. With this approach, simultaneous challenge of large cohorts of animals, gently restrained and not anesthetized, assures safe, well-controlled, unbiased, and uniform infection. In this chapter, we present a standardized method for reproducible aerosol delivery of wild-type Y. pestis to rodents for experimental models of primary pneumonic plague.


Subject(s)
Disease Models, Animal , Lung/microbiology , Plague/pathology , Pneumonia/pathology , Yersinia pestis/physiology , Aerosols/adverse effects , Animals , Humans , Lung/pathology , Mice , Plague/microbiology , Pneumonia/microbiology , Rats
7.
PLoS One ; 14(5): e0217440, 2019.
Article in English | MEDLINE | ID: mdl-31121001

ABSTRACT

Yersinia pestis is the causative agent of pneumonic plague, a disease involving uncontrolled bacterial growth and host immunopathology. Secondary septicemic plague commonly occurs as a consequence of the host inflammatory response that causes vasodilation and vascular leakage, which facilitates systemic spread of the bacteria and the colonization of secondary tissues. The mortality rates of pneumonic and septicemic plague are high even when antibiotics are administered. In this work, we show that primary pneumonic or secondary septicemic plague can be preferentially modeled in mice by varying the volume used for intranasal delivery of Y. pestis. Low volume intranasal challenge (10µL) of wild type Y. pestis resulted in a high frequency of lethal secondary septicemic plague, with a low degree of primary lung infection and rapid development of sepsis. In contrast, high volume intranasal challenge (30µL) yielded uniform early lung infection and primary disease and a significant increase in lethality. In a commonly used BSL2 model, high volume challenge with Y. pestis lacking the pigmentation locus (pgm-) gave 105-fold greater deposition compared to low volume challenge, yet moribund mice did not develop severe lung disease and there was no detectable difference in lethality. These data indicate the primary cause of death of mice in the BSL2 model is sepsis regardless of intranasal dosing method. Overall, these findings allow for the preferential modeling of pneumonic or septicemic plague by intranasal dosing of mice with Y. pestis.


Subject(s)
Plague/pathology , Pneumonia, Bacterial/pathology , Sepsis/pathology , Yersinia pestis/physiology , Animals , Disease Models, Animal , Female , Lung/microbiology , Lung/pathology , Male , Mice, Inbred C57BL , Plague/complications , Plague/microbiology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/microbiology , Sepsis/etiology , Sepsis/microbiology
8.
Infect Immun ; 87(4)2019 04.
Article in English | MEDLINE | ID: mdl-30642901

ABSTRACT

Yersinia pestis causes bubonic, pneumonic, and septicemic plague. Although no longer responsible for pandemic outbreaks, pneumonic plague continues to be a challenge for medical treatment and has been classified as a reemerging disease in some parts of the world. In the early stage of infection, inflammatory responses are believed to be suppressed by Y. pestis virulence factors in order to prevent clearance, while later, the hyperactivation of inflammation contributes to the progression of disease. In this work, we sought to identify the host factors that mediate this process and studied the role of the Toll/interleukin 1 (IL-1) receptor adapter and major inflammatory mediator myeloid differentiation primary response 88 (MyD88) in pneumonic plague. We show that pulmonary challenge of Myd88-/- mice with wild-type (WT) Y. pestis results in significant loss of pro- and anti-inflammatory cytokines and chemokines, especially gamma interferon (IFN-γ) and KC, in the lungs compared to that in WT mice. Bacterial growth in the lungs occurred more rapidly in the WT mice, however, indicating a role for the MyD88 response in facilitating the primary lung infection. Nevertheless, Myd88-/- mice were more sensitive to lethality from secondary septicemic plague. Together these findings indicate a central role for MyD88 during the biphasic inflammatory response to pulmonary Y. pestis infection. In the early phase, low-level MyD88-dependent chemokine expression limits initial growth but facilitates Y. pestis access to a protected replicative niche. The later hyperinflammatory phase is partially MyD88 dependent and ineffective in the lungs but controls systemic infection and reduces the progression of secondary septicemic plague.


Subject(s)
Lung/metabolism , Myeloid Differentiation Factor 88/metabolism , Plague/metabolism , Plague/microbiology , Yersinia pestis/growth & development , Animals , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Plague/genetics , Virulence , Yersinia pestis/genetics , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity
9.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28847850

ABSTRACT

Yersinia pestis causes bubonic, pneumonic, and septicemic plague, diseases that are rapidly lethal to most mammals, including humans. Plague develops as a consequence of bacterial neutralization of the host's innate immune response, which permits uncontrolled growth and causes the systemic hyperactivation of the inflammatory response. We previously found that host type I interferon (IFN) signaling is induced during Y. pestis infection and contributes to neutrophil depletion and disease. In this work, we show that type I IFN expression is derived from the recognition of intracellular Y. pestis by host Toll-like receptor 7 (TLR7). Type I IFN expression proceeded independent of myeloid differentiation factor 88 (MyD88), which is the only known signaling adaptor for TLR7, suggesting that a noncanonical mechanism occurs in Y. pestis-infected macrophages. In the murine plague model, TLR7 was a significant contributor to the expression of serum IFN-ß, whereas MyD88 was not. Furthermore, like the type I IFN response, TLR7 contributed to the lethality of septicemic plague and was associated with the suppression of neutrophilic inflammation. In contrast, TLR7 was important for defense against disease in the lungs. Together, these data demonstrate that an atypical TLR7 signaling pathway contributes to type I IFN expression during Y. pestis infection and suggest that the TLR7-driven type I IFN response plays an important role in determining the outcome of plague.


Subject(s)
Host-Pathogen Interactions , Interferon-beta/immunology , Membrane Glycoproteins/immunology , Myeloid Differentiation Factor 88/immunology , Plague/immunology , Toll-Like Receptor 7/immunology , Yersinia pestis/pathogenicity , Animals , Cell Line , Gene Expression Regulation , Immunity, Innate , Interferon-beta/genetics , Lung/immunology , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neutrophils/immunology , Neutrophils/microbiology , Plague/genetics , Plague/microbiology , Plague/mortality , Signal Transduction , Survival Analysis , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Virulence , Yersinia pestis/immunology
11.
J Lipid Res ; 58(3): 494-503, 2017 03.
Article in English | MEDLINE | ID: mdl-28053185

ABSTRACT

Unresolved experimental Lyme arthritis in C3H 5-lipoxygenase (5-LOX)-/- mice is associated with impaired macrophage phagocytosis of Borrelia burgdorferi In the present study, we further investigated the effects of the 5-LOX metabolite, leukotriene (LT)B4 on phagocytosis of B. burgdorferi Bone marrow-derived macrophages (BMDMs) from 5-LOX-/- mice were defective in the uptake and killing of B. burgdorferi from the earliest stages of spirochete internalization. BMDMs from mice deficient for the LTB4 high-affinity receptor (BLT1-/-) were also unable to efficiently phagocytose B. burgdorferi Addition of exogenous LTB4 augmented the phagocytic capability of BMDMs from both 5-LOX-/- and BLT1-/- mice, suggesting that the low-affinity LTB4 receptor, BLT2, might be involved. Blocking BLT2 activity with the specific antagonist, LY255283, inhibited phagocytosis in LTB4-stimulated BLT1-/- BMDMs, demonstrating a role for BLT2. However, the lack of a phagocytic defect in BLT2-/- BMDMs suggested that this was a compensatory effect. In contrast, 12(S)-hydroxyheptadeca-5Z,8E,10E-trienoic acid, a natural BLT2-specific high-affinity ligand, and resolvin E1, a BLT1 agonist, were both unable to boost phagocytosis in BMDMs from either 5-LOX-/- or BLT1-/- mice, suggesting a specific role for LTB4 in mediating phagocytosis in murine macrophages. This study demonstrates that LTB4 promotes macrophage phagocytosis of bacteria via BLT1, and that BLT2 can fulfill this role in the absence of BLT1.


Subject(s)
Arachidonate 5-Lipoxygenase/genetics , Lyme Disease/genetics , Receptors, Leukotriene B4/genetics , Animals , Arachidonate 5-Lipoxygenase/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Disease Models, Animal , Humans , Leukotriene B4/administration & dosage , Leukotriene B4/genetics , Leukotriene B4/metabolism , Lyme Disease/metabolism , Lyme Disease/microbiology , Lyme Disease/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Phagocytosis/genetics , Receptors, Leukotriene B4/antagonists & inhibitors , Tetrazoles/administration & dosage
12.
Methods Mol Biol ; 1531: 101-109, 2017.
Article in English | MEDLINE | ID: mdl-27837485

ABSTRACT

Many Gram-negative bacterial pathogens use type III secretion systems to export proteins that act directly on the host and aid in the infectious process. Extracellular bacteria primarily rely upon the type III secretion system to insert or inject effector proteins into the cytosol of their host cell in order to perturb intracellular signaling events and aid in pathogenesis. Intracellular bacteria can also depend on the T3SS translocation of effector proteins from vacuolar compartments into the vacuolar membrane or host cell cytosol where they can modulate intracellular trafficking and/or signaling pathways necessary for their growth and survival. Biochemical fractionation of infected cells in vitro enables detection of these events, making it possible to identify relevant protein-protein interactions, characterize phenotypes of mutant strains and understand how these effector proteins impact host cells. In this chapter we provide methods for the analysis of translocated effector proteins using biochemical and mechanical fractionation procedures.


Subject(s)
Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Cell Fractionation/methods , Bacterial Proteins/chemistry , Cell Line , Detergents , Gram-Negative Bacteria/metabolism , Protein Transport , Solubility , Subcellular Fractions , Type III Secretion Systems/metabolism , Vacuoles/metabolism
13.
Infect Immun ; 83(7): 2627-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870230

ABSTRACT

Infection of C3H mice with Borrelia burgdorferi, the causative agent of Lyme disease, reliably produces an infectious arthritis and carditis that peak around 3 weeks postinfection and then spontaneously resolve. Macrophage polarization has been suggested to drive inflammation, the clearance of bacteria, and tissue repair and resolution in a variety of infectious disease models. During Lyme disease it is clear that macrophages are capable of clearing Borrelia spirochetes and exhausted neutrophils; however, the role of macrophage phenotype in disease development or resolution has not been studied. Using classical (NOS2) and alternative (CD206) macrophage subset-specific markers, we determined the phenotype of F4/80(+) macrophages within the joints and heart throughout the infection time course. Within the joint, CD206(+) macrophages dominated throughout the course of infection, and NOS2(+) macrophage numbers became elevated only during the peak of inflammation. We also found dual NOS2(+) CD206(+) macrophages which increased during resolution. In contrast to findings for the ankle joints, numbers of NOS2(+) and CD206(+) macrophages in the heart were similar at the peak of inflammation. 5-Lipoxygenase-deficient (5-LOX(-/-)) mice, which display a failure of Lyme arthritis resolution, recruited fewer F4/80(+) cells to the infected joints and heart, but macrophage subset populations were unchanged. These results highlight differences in the inflammatory infiltrates during Lyme arthritis and carditis and demonstrate the coexistence of multiple macrophage subsets within a single inflammatory site.


Subject(s)
Borrelia burgdorferi/immunology , Cell Differentiation , Lyme Disease/immunology , Macrophages/immunology , Macrophages/physiology , Animals , Female , Immunophenotyping , Joints/pathology , Lectins, C-Type/analysis , Macrophages/chemistry , Mannose Receptor , Mannose-Binding Lectins/analysis , Mice, Inbred C3H , Myocardium/pathology , Nitric Oxide Synthase Type II/analysis , Receptors, Cell Surface/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...