Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Epidemiol ; 3: 1274800, 2023.
Article in English | MEDLINE | ID: mdl-38455907

ABSTRACT

Introduction: COVID-19 has emerged as a highly contagious and debilitating disease caused by the SARS-CoV-2 virus and has claimed the lives of over 7.7 million people worldwide. Bacterial co-infections are one of many co-morbidities that have been suggested to impact the outcome of COVID-19 in patients. The goals of this study are to elucidate the presence of bacteria in the nasopharynx of SARS-CoV-2 positive and negative patients and to describe demographic categories that may be associated with the detection of these organisms during one of the initial waves of the COVID-19 pandemic. Methods: To this end, we investigated SARS-CoV-2 and bacterial co-detection from outpatient RT-PCR testing in Texas. Results: The results indicate that Staphylococcus aureus, Streptococcus pneumoniae, Klebsiella pneumoniae, Moraxella catarrhalis, and Haemophilus influenzae were the most frequently detected bacteria in both SARS-CoV-2 positive and SARS-CoV-2 negative patients and that these bacteria were present in these two patient populations at similar proportions. We also detected Staphylococcus aureus in a significantly larger proportion of males relative to females and people under 65 years of age relative to those 65 and over. Finally, we observed that SARS-CoV-2 was more commonly detected in Hispanics compared to non-Hispanics; however, low disclosure rates make volunteer bias a concern when interpreting the effects of demographic variables. Discussion: This study describes the bacteria present in the nasopharynx of SARS-CoV-2 positive and negative patients, highlights associations between patient demographics and SARS-CoV-2 as well as bacterial co-detection. In addition, this study highlights RT-PCR based molecular testing as a tool to detect bacteria simultaneously when SARS-CoV-2 tests are performed.

2.
PLoS Pathog ; 18(12): e1011020, 2022 12.
Article in English | MEDLINE | ID: mdl-36542660

ABSTRACT

BACKGROUND: For almost a century, it has been recognized that influenza A virus (IAV) infection can promote the development of secondary bacterial infections (SBI) mainly caused by Streptococcus pneumoniae (Spn). Recent observations have shown that IAV is able to directly bind to the surface of Spn. To gain a foundational understanding of how direct IAV-Spn interaction alters bacterial biological fitness we employed combinatorial multiomic and molecular approaches. RESULTS: Here we show IAV significantly remodels the global transcriptome, proteome and phosphoproteome profiles of Spn independently of host effectors. We identified Spn surface proteins that interact with IAV proteins (hemagglutinin, nucleoprotein, and neuraminidase). In addition, IAV was found to directly modulate expression of Spn virulence determinants such as pneumococcal surface protein A, pneumolysin, and factors associated with antimicrobial resistance among many others. Metabolic pathways were significantly altered leading to changes in Spn growth rate. IAV was also found to drive Spn capsule shedding and the release of pneumococcal surface proteins. Released proteins were found to be involved in evasion of innate immune responses and actively reduced human complement hemolytic and opsonizing activity. IAV also led to phosphorylation changes in Spn proteins associated with metabolism and bacterial virulence. Validation of proteomic data showed significant changes in Spn galactose and glucose metabolism. Furthermore, supplementation with galactose rescued bacterial growth and promoted bacterial invasion, while glucose supplementation led to enhanced pneumolysin production and lung cell apoptosis. CONCLUSIONS: Here we demonstrate that IAV can directly modulate Spn biology without the requirement of host effectors and support the notion that inter-kingdom interactions between human viruses and commensal pathobionts can promote bacterial pathogenesis and microbiome dysbiosis.


Subject(s)
Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Humans , Streptococcus pneumoniae/metabolism , Influenza A virus/genetics , Virulence , Galactose/metabolism , Multiomics , Proteomics , Influenza, Human/genetics , Influenza, Human/complications
3.
Infect Immun ; 89(12): e0043021, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34543120

ABSTRACT

Despite the maintenance of YopP/J alleles throughout the human-pathogenic Yersinia lineage, the benefit of YopP/J-induced phagocyte death for Yersinia pathogenesis in animals is not obvious. To determine how the sequence divergence of YopP/J has impacted Yersinia virulence, we examined protein polymorphisms in this type III secreted effector protein across 17 Yersinia species and tested the consequences of polymorphism in a murine model of subacute systemic yersiniosis. Our evolutionary analysis revealed that codon 177 has been subjected to positive selection; the Yersinia enterocolitica residue had been altered from a leucine to a phenylalanine in nearly all Yersinia pseudotuberculosis and Yersinia pestis strains examined. Despite this change being minor, as both leucine and phenylalanine have hydrophobic side chains, reversion of YopJF177 to the ancestral YopJL177 variant yielded a Y. pseudotuberculosis strain with enhanced cytotoxicity toward macrophages, consistent with previous findings. Surprisingly, expression of YopJF177L in the mildly attenuated ksgA- background rendered the strain completely avirulent in mice. Consistent with this hypothesis that YopJ activity relates indirectly to Yersinia pathogenesis in vivo, ksgA- strains lacking functional YopJ failed to kill macrophages but actually regained virulence in animals. Also, treatment with the antiapoptosis drug suramin prevented YopJ-mediated macrophage cytotoxicity and enhanced Y. pseudotuberculosis virulence in vivo. Our results demonstrate that Yersinia-induced cell death is detrimental for bacterial pathogenesis in this animal model of illness and indicate that positive selection has driven YopJ/P and Yersinia evolution toward diminished cytotoxicity and increased virulence, respectively.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Yersinia Infections/microbiology , Yersinia/physiology , Animals , Bacterial Proteins/metabolism , Disease Susceptibility , Humans , Mutation , Virulence/genetics , Virulence Factors , Yersinia/pathogenicity
4.
PLoS Negl Trop Dis ; 10(6): e0004787, 2016 06.
Article in English | MEDLINE | ID: mdl-27332553

ABSTRACT

Neurocysticercosis (NCC) is one of the most common helminth parasitic diseases of the central nervous system (CNS) and the leading cause of acquired epilepsy worldwide. NCC is caused by the presence of the metacestode larvae of the tapeworm Taenia solium within brain tissues. NCC patients exhibit a long asymptomatic phase followed by a phase of symptoms including increased intra-cranial pressure and seizures. While the asymptomatic phase is attributed to the immunosuppressive capabilities of viable T. solium parasites, release of antigens by dying organisms induce strong immune responses and associated symptoms. Previous studies in T. solium-infected pigs have shown that the inflammatory response consists of various leukocyte populations including eosinophils, macrophages, and T cells among others. Because the role of eosinophils within the brain has not been investigated during NCC, we examined parasite burden, disease susceptibility and the composition of the inflammatory reaction in the brains of infected wild type (WT) and eosinophil-deficient mice (ΔdblGATA) using a murine model of NCC in which mice were infected intracranially with Mesocestoides corti, a cestode parasite related to T. solium. In WT mice, we observed a time-dependent induction of eosinophil recruitment in infected mice, contrasting with an overall reduced leukocyte infiltration in ΔdblGATA brains. Although, ΔdblGATA mice exhibited an increased parasite burden, reduced tissue damage and less disease susceptibility was observed when compared to infected WT mice. Cellular infiltrates in infected ΔdblGATA mice were comprised of more mast cells, and αß T cells, which correlated with an abundant CD8+ T cell response and reduced CD4+ Th1 and Th2 responses. Thus, our data suggest that enhanced inflammatory response in WT mice appears detrimental and associates with increased disease susceptibility, despite the reduced parasite burden in the CNS. Overall reduced leukocyte infiltration due to absence of eosinophils correlates with attenuated tissue damage and longer survival of ΔdblGATA mice. Therefore, our study suggests that approaches to clear NCC will require strategies to tightly control the host immune response while eradicating the parasite with minimal damage to brain tissue.


Subject(s)
Eosinophilia/genetics , Genetic Predisposition to Disease , Leukocytes/physiology , Neurocysticercosis/pathology , Animals , Female , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurocysticercosis/genetics , Neutrophils/physiology
5.
Infect Immun ; 83(2): 614-24, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422267

ABSTRACT

Serratia marcescens, a member of the carbapenem-resistant Enterobacteriaceae, is an important emerging pathogen that causes a wide variety of nosocomial infections, spreads rapidly within hospitals, and has a systemic mortality rate of ≤41%. Despite multiple clinical descriptions of S. marcescens nosocomial pneumonia, little is known regarding the mechanisms of bacterial pathogenesis and the host immune response. To address this gap, we developed an oropharyngeal aspiration model of lethal and sublethal S. marcescens pneumonia in BALB/c mice and extensively characterized the latter. Lethal challenge (>4.0 × 10(6) CFU) was characterized by fulminate hemorrhagic pneumonia with rapid loss of lung function and death. Mice challenged with a sublethal dose (<2.0 × 10(6) CFU) rapidly lost weight, had diminished lung compliance, experienced lung hemorrhage, and responded to the infection with extensive neutrophil infiltration and histopathological changes in tissue architecture. Neutrophil extracellular trap formation and the expression of inflammatory cytokines occurred early after infection. Mice depleted of neutrophils were exquisitely susceptible to an otherwise nonlethal inoculum, thereby demonstrating the requirement for neutrophils in host protection. Mutation of the genes encoding the cytolysin ShlA and its transporter ShlB resulted in attenuated S. marcescens strains that failed to cause profound weight loss, extended illness, hemorrhage, and prolonged lung pathology in mice. This study describes a model of S. marcescens pneumonia that mimics known clinical features of human illness, identifies neutrophils and the toxin ShlA as a key factors important for defense and infection, respectively, and provides a solid foundation for future studies of novel therapeutics for this important opportunistic pathogen.


Subject(s)
Bacterial Proteins/genetics , Hemolysin Proteins/genetics , Pneumonia/pathology , Serratia Infections/immunology , Serratia marcescens/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cross Infection , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Female , Hemorrhage/microbiology , Hemorrhage/pathology , Inflammation/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/immunology , Neutrophils/immunology , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/mortality , Serratia Infections/microbiology , Serratia Infections/mortality , Serratia marcescens/pathogenicity
6.
Microb Pathog ; 51(1-2): 39-47, 2011.
Article in English | MEDLINE | ID: mdl-21440052

ABSTRACT

The virulence mechanisms of Francisella tularensis, the causative agent of severe pneumonia in humans and a CDC category A bioterrorism agent, are not fully defined. As sepsis is the leading cause of mortality associated with respiratory infections, we determined whether, in the absence of any known bacterial toxins, a deregulated host response resulting in sepsis syndrome is associated with lethality of respiratory infection with the virulent human Type A strain SchuS4 of F. tularensis. The C57BL/6 mice infected intranasally with a lethal dose of SchuS4 exhibited high bacterial burden in systemic organs and blood indicative of bacteremia. In correlation, infected mice displayed severe tissue pathology and associated cell death in lungs, liver and spleen. Consistent with our studies with murine model strain Francisella novicida, infection with SchuS4 caused an initial delay in upregulation of inflammatory mediators followed by development of severe sepsis characterized by exaggerated cytokine release, upregulation of cardiovascular injury markers and sepsis mediator alarmins S100A9 and HMGB1. This study shows that pulmonary tularemia caused by the Type A strain of F. tularensis results in a deregulated host response leading to severe sepsis and likely represents the major cause of mortality associated with this virulent pathogen.


Subject(s)
Francisella tularensis/pathogenicity , Lung Diseases/complications , Sepsis/pathology , Tularemia/complications , Animals , Bacteremia/pathology , Cytokines/blood , Humans , Inflammation , Liver/microbiology , Liver/pathology , Lung/microbiology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Sepsis/etiology , Sepsis/immunology , Sepsis/microbiology , Spleen/microbiology , Spleen/pathology , Tularemia/immunology , Tularemia/microbiology , Tularemia/pathology , Up-Regulation , Virulence
7.
Immunol Cell Biol ; 89(2): 167-72, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20585334

ABSTRACT

The macrophage is a versatile cell type that can sense and respond to a particular need based on the conditions of the microenvironment. Some studies have recently suggested that pathogens can directly influence the polarization of macrophages. As Francisella infections are characterized by intense necrotic infiltrates in the lung as well as in distal sites of infection, we sought to investigate whether pulmonary Francisella infections could cause the polarization of alternatively activated macrophages (M2/aaMs). Our results indicate that Francisella infections can cause the polarization of M2/aaM in vivo and that macrophages can be polarized toward an M2/aaM phenotype more potently if dead cell debris is used for stimulation in the presence and absence of Francisella infections. Finally, we also demonstrate that efferocytosis is inhibited in macrophages infected with Francisella, thus providing a potential explanation for the lack of clearance and eventual accumulation of dead cell debris associated with this disease.


Subject(s)
Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/microbiology , Phagocytosis/immunology , Animals , Arginase/biosynthesis , Biomarkers , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Models, Immunological , Necrosis , Up-Regulation
8.
PLoS One ; 5(11): e14088, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21124895

ABSTRACT

BACKGROUND: Pneumonia and pulmonary infections are major causes of mortality among the growing elderly population. Age associated attenuations of various immune parameters, involved with both innate and adaptive responses are collectively known as immune senescence. These changes are likely to be involved with differences in host susceptibility to disease between young and aged individuals. METHODOLOGY/PRINCIPAL FINDINGS: The objective of this study was to assess potential age related differences in the pulmonary host response in mice to the Gram-negative respiratory pathogen, Francisella novicida. We intranasally infected mice with F. novicida and compared various immune and pathological parameters of the pulmonary host response in both young and aged mice. CONCLUSIONS/SIGNIFICANCE: We observed that 20% of aged mice were able to survive an intranasal challenge with F. novicida while all of their younger cohorts died consistently within 4 to 6 days post infection. Further experiments revealed that all of the aged mice tested were initially able to control bacterial replication in the lungs as well as at distal sites of replication compared with young mice. In addition, the small cohort of aged survivors did not progress to a severe sepsis syndrome with hypercytokinemia, as did all of the young adult mice. Finally, a lack of widespread cell death in potential aged survivors coupled with a difference in cell types recruited to sites of infection within the lung confirmed an altered host response to Francisella in aged mice.


Subject(s)
Aging/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Lung Diseases/immunology , Animals , Apoptosis/immunology , Cell Survival/immunology , Cytokines/blood , Cytokines/immunology , Female , Francisella/physiology , Gram-Negative Bacterial Infections/microbiology , Host-Pathogen Interactions/immunology , Inflammation Mediators/blood , Inflammation Mediators/immunology , Kaplan-Meier Estimate , Lung/immunology , Lung/microbiology , Lung/pathology , Lung Diseases/microbiology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
9.
BMC Microbiol ; 8: 215, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19068128

ABSTRACT

BACKGROUND: Francisella tularensis subsp. tularensis is classified as a Category A bioweapon that is capable of establishing a lethal infection in humans upon inhalation of very few organisms. However, the virulence mechanisms of this organism are not well characterized. Francisella tularensis subsp. novicida, which is an equally virulent subspecies in mice, was used in concert with a microPET scanner to better understand its temporal dissemination in vivo upon intranasal infection and how such dissemination compares with other routes of infection. Adult mice were inoculated intranasally with F. tularensis subsp. novicida radiolabeled with 64Cu and imaged by microPET at 0.25, 2 and 20 hours post-infection. RESULTS: 64Cu labeled F. tularensis subsp. novicida administered intranasally or intratracheally were visualized in the respiratory tract and stomach at 0.25 hours post infection. By 20 hours, there was significant tropism to the lung compared with other tissues. In contrast, the images of radiolabeled F. tularensis subsp. novicida when administered intragastrically, intradermally, intraperitoneally and intravenouslly were more generally limited to the gastrointestinal system, site of inoculation, liver and spleen respectively. MicroPET images correlated with the biodistribution of isotope and bacterial burdens in analyzed tissues. CONCLUSION: Our findings suggest that Francisella has a differential tissue tropism depending on the route of entry and that the virulence of Francisella by the pulmonary route is associated with a rapid bacteremia and an early preferential tropism to the lung. In addition, the use of the microPET device allowed us to identify the cecum as a novel site of colonization of Francisella tularensis subsp. novicida in mice.


Subject(s)
Francisella tularensis/pathogenicity , Tularemia/microbiology , Tularemia/pathology , Animals , Copper Radioisotopes/analysis , Francisella tularensis/isolation & purification , Gastrointestinal Tract/microbiology , Liver/microbiology , Lung/microbiology , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods , Spleen/microbiology , Staining and Labeling , Whole Body Imaging/methods
10.
Infect Immun ; 76(7): 3001-10, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18411294

ABSTRACT

"Francisella tularensis subsp. novicida" intranasal infection causes a rapid pneumonia in mice with mortality at 4 to 6 days with a low dose of bacteria (10(2) bacteria). The short time to death suggests that there is a failure of the innate immune response. As the neutrophil is often the first cell type to infiltrate sites of infection, we focused on the emigration of neutrophils in this infection, as well as cytokines involved in their recruitment. The results indicated that there was a significant delay in the influx of neutrophils into the bronchoalveolar lavage fluid of F. tularensis subsp. novicida-infected mice. The delay in neutrophil recruitment in F. tularensis subsp. novicida-infected mice correlated with a delay in the upregulation of multiple proinflammatory cytokines and chemokines, as well as a delay in caspase-1 activation. Strikingly, the initial delay in the upregulation of cytokines through 1 day postinfection was followed by profound upregulation of multiple cytokines and chemokines to levels consistent with hypercytokinemia described for severe sepsis. This finding was further supported by a bacteremia and the cellular relocalization and release of high-mobility group box-1 and S100A9, both of which are damage-associated molecular pattern molecules and are known to be mediators of severe sepsis.


Subject(s)
Bacteremia , Calgranulin B/metabolism , Cytokines/metabolism , Francisella tularensis/pathogenicity , HMGB1 Protein/metabolism , Pneumonia, Bacterial , Up-Regulation , Animals , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/physiopathology , Blood/microbiology , Cell Line , Colony Count, Microbial , Female , Francisella tularensis/immunology , Francisella tularensis/isolation & purification , Humans , Lung/immunology , Lung/microbiology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/physiopathology , Time Factors , Tularemia/immunology , Tularemia/microbiology , Tularemia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...