Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 330
Filter
1.
J Infect Dis ; 223(2): 333-341, 2021 02 03.
Article in English | MEDLINE | ID: mdl-32572481

ABSTRACT

BACKGROUND: Otitis media (OM) is a common and potentially serious disease of childhood. Although OM is multifactorial on origin, bacterial infection is a unifying component. Many studies have established a critical role for innate immunity in bacterial clearance and OM resolution. A key component of innate immunity is the recruitment of immune and inflammatory cells, including macrophages. METHODS: To explore the role of macrophages in OM, we evaluated the expression of genes related to macrophage function during a complete episode of acute OM in the mouse caused by middle ear (ME) inoculation with Haemophilus influenzae. We also combined CCR2 deficiency with chlodronate liposome toxicity to deplete macrophages during OM. RESULTS: Macrophage genes were robustly regulated during OM. Moreover, macrophage depletion enhanced and prolonged the infiltration of neutrophils into the infected ME and increased the persistence of bacterial infection. CONCLUSIONS: The results illustrate the critical role played by macrophages in OM resolution.


Subject(s)
Bacterial Infections/etiology , Macrophages/immunology , Macrophages/metabolism , Neutrophil Infiltration/immunology , Otitis Media/etiology , Receptors, CCR2/deficiency , Animals , Bacterial Infections/metabolism , Bacterial Infections/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Haemophilus Infections/etiology , Haemophilus Infections/pathology , Haemophilus influenzae/immunology , Mice , Mice, Knockout , Otitis Media/pathology
2.
Proc Natl Acad Sci U S A ; 115(30): E7149-E7157, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29987031

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a major cause of community acquired pneumonia and exacerbation of chronic obstructive pulmonary disease. A current effort in NTHi vaccine development has focused on generating humoral responses and has been greatly impeded by antigenic variation among the numerous circulating NTHi strains. In this study, we showed that pulmonary immunization of mice with killed NTHi generated broad protection against lung infection by different strains. While passive transfer of immune antibodies protected only against the homologous strain, transfer of immune T cells conferred protection against both homologous and heterologous strains. Further characterization revealed a strong Th17 response that was cross-reactive with different NTHi strains. Responding Th17 cells recognized both cytosolic and membrane-associated antigens, while immune antibodies preferentially responded to surface antigens and were highly strain specific. We further identified several conserved proteins recognized by lung Th17 cells during NTHi infection. Two proteins yielding the strongest responses were tested as vaccine candidates by immunization of mice with purified proteins plus an adjuvant. Immunization induced antigen-specific Th17 cells that recognized different strains and, upon adoptive transfer, conferred protection. Furthermore, immunized mice were protected against challenge with not only NTHi strains but also a fully virulent, encapsulated strain. Together, these results show that the immune mechanism of cross-protection against pneumonia involves Th17 cells, which respond to a broad spectrum of antigens, including those that are highly conserved among NTHi strains. These mechanistic insights suggest that inclusion of Th17 antigens in subunit vaccines offers the advantage of inducing broad protection and complements the current antibody-based approaches.


Subject(s)
Antigens, Bacterial/immunology , Haemophilus Infections/immunology , Haemophilus Vaccines/immunology , Haemophilus influenzae/immunology , Pneumonia, Bacterial/immunology , Th17 Cells/immunology , Animals , Cross Reactions , Haemophilus Infections/pathology , Haemophilus Infections/prevention & control , Mice , Mice, Knockout , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/prevention & control , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/prevention & control , Th17 Cells/pathology
3.
Microb Pathog ; 141: 103985, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31968224

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a common airway commensal and opportunistic pathogen that persists within biofilm communities in vivo. Biofilm studies so far are mainly based on assays on plastic surfaces. The aim of this work was to investigate the capacity of clinical NTHi strains to form biofilm structures on polarized Calu-3 human airway epithelial cells and primary normal human bronchial epithelial cells and to characterize the biofilm architecture. Formation of adherent NTHi biofilms post colonization of host cells at multiple time-points was evaluated using confocal laser scanning microscopy and electron microscopy. NTHi biofilms were analyzed in terms of biofilm height and presence of extracellular matrix components, and their apoptotic effects on epithelial cells were measured by TUNEL assay. Strain Fi176 was observed to form robust biofilms on airway epithelia over time, while disrupting the integrity of Calu-3 monolayer by 72 h of co-culture. NTHi biofilms were observed to induce apoptotic DNA fragmentation in host cells at 24 h post infection. Biofilm formation on cell monolayers by Fi176ΔpilA strain was markedly reduced compared to WT strain. Biofilm inhibition and disruption assays by crystal violet staining indicated that DNA and proteins are part of NTHi biofilms in vitro. Our findings highlight critical stages of NTHi pathogenesis following host colonization and provide useful biofilm models for future antimicrobial drug discovery investigations.


Subject(s)
Biofilms , DNA Fragmentation , DNA, Bacterial , Haemophilus Infections/microbiology , Haemophilus influenzae/growth & development , Haemophilus influenzae/genetics , Respiratory Mucosa/microbiology , Apoptosis , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae/classification , Haemophilus influenzae/ultrastructure , Humans , Respiratory Mucosa/pathology
4.
Respir Res ; 21(1): 170, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32620122

ABSTRACT

Asthma is a complex heterogeneous disease. The neutrophilic subtypes of asthma are described as persistent, more severe and corticosteroid-resistant, with higher hospitalization and mortality rates, which seriously affect the lives of asthmatic patients. With the development of high-throughput sequencing technology, an increasing amount of evidence has shown that lower airway microbiome dysbiosis contributes to the exacerbation of asthma, especially neutrophilic asthma. Nontypeable Haemophilus influenzae is normally found in the upper respiratory tract of healthy adults and is one of the most common strains in the lower respiratory tract of neutrophilic asthma patients, in whom its presence is related to the occurrence of corticosteroid resistance. To understand the pathogenic mechanism by which nontypeable Haemophilus influenzae colonization leads to the progression of neutrophilic asthma, we reviewed the previous literature on nontypeable Haemophilus influenzae colonization and subsequent aggravation of neutrophilic asthma and corticosteroid resistance. We discussed nontypeable Haemophilus influenzae as a potential therapeutic target to prevent the progression of neutrophilic asthma.


Subject(s)
Asthma/microbiology , Asthma/pathology , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae , Neutrophils/pathology , Humans
5.
Cell Microbiol ; 21(1): e12960, 2019 01.
Article in English | MEDLINE | ID: mdl-30265765

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) is a major pathogen causing acute otitis media (AOM). The relationship between the cellular content of the middle ear fluid (MEF) during AOM and infection of NTHi is poorly understood. Using the Junbo mouse, a characterised NTHi infection model, we analysed the cellular content of MEF and correlated the data with NTHi titres. The MEF of the Junbo mouse was heterogeneous between ears and was graded from 1 to 5; 1 being highly serous/clear and 5 being heavily viscous/opaque. At seven-day post-intranasal inoculation, NTHi was not found in grade-1 or 2 fluids, and the proportion of MEF that supported NTHi increased with the grade. Analyses by flow cytometry indicated that the cellular content was highest in grade-4 and 5 fluids, with a greater proportion of necrotic cells and a low-live cell count. NTHi infection of the middle ear increased the cell count and led to infiltration of immune cells and changes in the cytokine and chemokine levels. Following NTHi inoculation, high-grade infected MEFs had greater neutrophil infiltration whereas monocyte infiltration was significantly higher in serous noninfected low-grade fluids. These data underline a role for immune cells, specifically monocytes and neutrophils, and cell necrosis in NTHi infection of the Junbo mouse middle ear.


Subject(s)
Ear, Middle/microbiology , Ear, Middle/pathology , Exudates and Transudates/cytology , Haemophilus Infections/pathology , Haemophilus influenzae/growth & development , Otitis Media/pathology , Animals , Cell Count , Disease Models, Animal , Flow Cytometry , Mice , Monocytes/immunology , Neutrophils/immunology
6.
Infection ; 48(2): 285-288, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31549360

ABSTRACT

OBJECTIVES: There is increasing evidence indicating an association between invasive non-typeable Haemophilus influenzae (NTHi) infection in pregnancy and early pregnancy loss. As the diagnosis relies on microbiological investigation of post-mortem placental and foetal samples, a significant proportion of NTHi-related pregnancy loss remains unrecognised. To better characterise NTHi in septic abortion, we report NTHi cases associated with early pregnancy loss. METHODS: We reviewed all post-mortems at <24 weeks gestation with histologically proven acute chorioamnionitis on placental histology and enrolled cases with at least one matched foetal and placental sample culture positive for NTHi. The study was approved by the NHS Lothian Caldicott Guardian. RESULTS: In our cohort, invasive NTHi has accounted for 20% of infections associated with early pregnancy loss prior to 24 weeks gestation. All patients were young and healthy pregnant women at < 20 weeks' gestation who presented with abdominal pain, PV bleed /discharge and were septic at the time of presentation. One patient with previous history of miscarriage who presented with cervical incompetence had more severe pathology suggestive of early intrauterine pneumonia. CONCLUSION: The burden of invasive NTHi disease in early pregnancy loss is likely to be much larger than currently recognised. NTHi should be considered in pregnant women presenting with abdominal pain and PV bleed/discharge in whom clinical signs of sepsis are present. Active surveillance should be considered in this patient group including septic abortion to capture the true prevalence of this emerging pathogen to inform preventative and therapeutic approaches.


Subject(s)
Abortion, Spontaneous/etiology , Communicable Diseases, Emerging/complications , Communicable Diseases, Emerging/microbiology , Haemophilus Infections/complications , Adult , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/pathology , Female , Genotype , Haemophilus Infections/drug therapy , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae/classification , Haemophilus influenzae/genetics , Humans , Scotland , Sepsis/drug therapy , Sepsis/microbiology
7.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30833337

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a major human pathogen, responsible for several acute and chronic infections of the respiratory tract. The incidence of invasive infections caused by NTHi is increasing worldwide. NTHi is able to colonize the nasopharynx asymptomatically, and the exact change(s) responsible for transition from benign carriage to overt disease is not understood. We have previously reported that phase variation (the rapid and reversible ON-OFF switching of gene expression) of particular lipooligosaccharide (LOS) glycosyltransferases occurs during transition from colonizing the nasopharynx to invading the middle ear. Variation in the structure of the LOS is dependent on the ON/OFF expression status of each of the glycosyltransferases responsible for LOS biosynthesis. In this study, we surveyed a collection of invasive NTHi isolates for ON/OFF expression status of seven phase-variable LOS glycosyltransferases. We report that the expression state of the LOS biosynthetic genes oafA ON and lic2A OFF shows a correlation with invasive NTHi isolates. We hypothesize that these gene expression changes contribute to the invasive potential of NTHi. OafA expression, which is responsible for the addition of an O-acetyl group onto the LOS, has been shown to impart a phenotype of increased serum resistance and may serve as a marker for invasive NTHi.


Subject(s)
Haemophilus Infections/genetics , Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Haemophilus influenzae/pathogenicity , Host-Pathogen Interactions/immunology , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/genetics , Lipopolysaccharides/immunology , Haemophilus Infections/pathology , Haemophilus influenzae/genetics , Host-Pathogen Interactions/genetics , Humans , Queensland
8.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31548315

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a major pathogen causing acute otitis media (AOM). The pathology of AOM increases during long-term infection in the middle ear (ME), but the host cellular immune response to bacterial infection in this inflamed environment is poorly understood. Using the Junbo mouse, a characterized NTHi infection model, we analyzed the cellular response to NTHi infection in the Junbo mouse middle ear fluid (MEF). NTHi infection increased the total cell number and significantly decreased the proportion of live cells in the MEF at day 1, and this further decreased gradually on each day up to day 7. Flow cytometry analysis showed that neutrophils were the dominant immune cell population in the MEF and that NTHi infection significantly increased their proportion whereas it decreased the monocyte, macrophage, and dendritic cell proportions. Neutrophil and macrophage numbers increased in blood and spleen after NTHi infection. The T-cell population was dominated by T-helper (Th) cells in noninoculated MEF, and the effector Th (CD44+) cell population increased at day 2 of NTHi infection with an increase in IL-12p40 levels. Sustained NTHi infection up to 3 days increased the transforming growth factor ß levels, decreasing the effector cell population and increasing the T-regulatory (T-reg) cell population. In the preinflamed ME environment of the Junbo mouse, neutrophils are the first responder to NTHi infection followed by T-reg immune suppressive cells. These data indicate that sustained NTHi infection in the ME induces the immune suppressive response by inducing the T-reg cell population and reducing immune cell infiltration, thus promoting longer-term infection.


Subject(s)
Ear, Middle/pathology , Haemophilus Infections/pathology , Haemophilus influenzae/immunology , Neutrophils/immunology , Otitis Media with Effusion/pathology , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Ear, Middle/microbiology , Haemophilus Infections/microbiology , Interleukin-12 Subunit p40/metabolism , Macrophages/immunology , Mice , Otitis Media with Effusion/microbiology , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta1/metabolism
9.
Int J Med Microbiol ; 308(7): 829-839, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30049648

ABSTRACT

The Gram-negative bacterium Haemophilus influenzae (H. influenzae) can commensally colonize the upper respiratory tract, but also cause life threatening disease including epiglottitis, sepsis and meningitis. The H. influenzae capsule protects the bacteria against both phagocytosis and opsonization. Encapsulated H. influenzae strains are classified into serotypes ranging from a to f dependent on their distinct polysaccharide capsule. Due to the implementation of vaccination the incidence of invasive H. influenzae type b (Hib) infections has strongly decreased and infections with other capsulated types, including H. influenzae type f (Hif), are emerging. The pathogenesis of H. influenzae meningitis is not clarified. To enter the central nervous system (CNS) the bacteria generally have to cross either the blood-brain barrier (BBB) or the blood-cerebrospinal fluid barrier (BSCFB). Using a cell culture model of the BCSFB based on human choroid plexus papilloma (HIBCPP) cells and different H. influenzae strains we investigated whether Hib and Hif invade the cells, and if invasion differs between encapsulated vs. capsular-deficient and fimbriated vs. non-fimbriated variants. We find that Hib can adhere to and invade into HIBCPP cells. Invasion occurs in a strongly polar fashion, since the bacteria enter the cells preferentially from the basolateral "blood "side. Fimbriae and capsule attenuate invasion into choroid plexus (CP) epithelial cells, and capsulation can influence the bacterial distribution pattern. Finally, analysis of clinical Hib and Hif isolates confirms the detected invasive properties of H. influenzae. Our data point to roles of capsule and fimbriae during invasion of CP epithelial cells.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Capsules/pathology , Blood-Brain Barrier/microbiology , Fimbriae, Bacterial/pathology , Haemophilus Infections/pathology , Haemophilus influenzae/pathogenicity , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Haemophilus influenzae/classification , Haemophilus influenzae/isolation & purification , Host-Pathogen Interactions/physiology , Humans
10.
Crit Rev Microbiol ; 44(2): 125-142, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28539074

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is a leading cause of morbidity and mortality worldwide. In the lower airways of COPD patients, bacterial infection is a common phenomenon and Haemophilus influenzae is the most commonly identified bacteria. Haemophilus influenzae is divided into typeable and nontypeable (NTHi) strains based on the presence or absence of a polysaccharide capsule. While NTHi is a common commensal in the human nasopharynx, it is associated with considerable inflammation when it is present in the lower airways of COPD patients, resulting in morbidity due to worsening symptoms and increased frequency of COPD exacerbations. Treatment of lower airway NTHi infection with antibiotics, though successful in the short term, does not offer long-term protection against reinfection, nor does it change the course of the disease. Hence, there has been much interest in the development of an effective NTHi vaccine. This review will summarize the current literature concerning the role of NTHi infections in COPD patients and the consequences of using prophylactic antibiotics in patients with COPD. There is particular focus on the rationale, findings of clinical studies and possible future directions of NTHi vaccines in patients with COPD.


Subject(s)
Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae/classification , Haemophilus influenzae/isolation & purification , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/pathology , Anti-Bacterial Agents/therapeutic use , Haemophilus Infections/drug therapy , Haemophilus Infections/prevention & control , Haemophilus Vaccines/immunology , Haemophilus Vaccines/isolation & purification , Humans
11.
Infect Immun ; 85(9)2017 09.
Article in English | MEDLINE | ID: mdl-28674033

ABSTRACT

Haemophilus parainfluenzae is a nutritionally fastidious, Gram-negative bacterium with an oropharyngeal/nasopharyngeal carriage niche that is associated with a range of opportunistic infections, including infectious endocarditis and otitis media (OM). These infections are often chronic/recurrent in nature and typically involve bacterial persistence within biofilm communities that are highly resistant to host clearance. This study addresses the primary hypothesis that H. parainfluenzae forms biofilm communities that are important determinants of persistence in vivo The results from in vitro biofilm studies confirmed that H. parainfluenzae formed biofilm communities within which the polymeric matrix was mainly composed of extracellular DNA and proteins. Using a chinchilla OM infection model, we demonstrated that H. parainfluenzae formed surface-associated biofilm communities containing bacterial and host components that included neutrophil extracellular trap (NET) structures and that the bacteria mainly persisted in these biofilm communities. We also used this model to examine the possible interaction between H. parainfluenzae and its close relative Haemophilus influenzae, which is also commonly carried within the same host environments and can cause OM. The results showed that coinfection with H. influenzae promoted clearance of H. parainfluenzae from biofilm communities during OM infection. The underlying mechanisms for bacterial persistence and biofilm formation by H. parainfluenzae and knowledge about the survival defects of H. parainfluenzae during coinfection with H. influenzae are topics for future work.


Subject(s)
Biofilms/growth & development , Haemophilus Infections/microbiology , Haemophilus parainfluenzae/physiology , Otitis Media/microbiology , Animals , Antibiosis , Chinchilla , Disease Models, Animal , Haemophilus Infections/pathology , Haemophilus influenzae/growth & development , Haemophilus influenzae/physiology , Haemophilus parainfluenzae/growth & development , Otitis Media/pathology
12.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28847849

ABSTRACT

We previously found CC chemokine ligand 3 (CCL3) to be a potent effector of inflammation during otitis media (OM): exogenous CCL3 rescues the OM phenotype of tumor necrosis factor-deficient mice and the function of macrophages deficient in several innate immune molecules. To further delineate the role of CCL3 in OM, we evaluated middle ear (ME) responses of ccl3-/-mice to nontypeable Haemophilus influenzae (NTHi). CCL chemokine gene expression was evaluated in wild-type (WT) mice during the complete course of acute OM. OM was induced in ccl3-/- and WT mice, and infection and inflammation were monitored for 21 days. Phagocytosis and killing of NTHi by macrophages were evaluated by an in vitro assay. The nasopharyngeal bacterial load was assessed in naive animals of both strains. Many CCL genes showed increased expression levels during acute OM, with CCL3 being the most upregulated, at levels 600-fold higher than the baseline. ccl3-/- deletion compromised ME bacterial clearance and prolonged mucosal hyperplasia. ME recruitment of leukocytes was delayed but persisted far longer than in WT mice. These events were linked to a decrease in the macrophage capacity for NTHi phagocytosis and increased nasopharyngeal bacterial loads in ccl3-/- mice. The generalized impairment in inflammatory cell recruitment was associated with compensatory changes in the expression profiles of CCL2, CCL7, and CCL12. CCL3 plays a significant role in the clearance of infection and resolution of inflammation and contributes to mucosal host defense of the nasopharyngeal niche, a reservoir for ME and upper respiratory infections. Therapies based on CCL3 could prove useful in treating or preventing persistent disease.


Subject(s)
Chemokine CCL3/immunology , Ear, Middle/immunology , Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Nasopharynx/immunology , Otitis Media/immunology , Animals , Bacterial Load , Cell Movement , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL3/deficiency , Chemokine CCL3/genetics , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Disease Models, Animal , Ear, Middle/microbiology , Gene Expression Regulation , Haemophilus Infections/genetics , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Host-Pathogen Interactions , Leukocytes/immunology , Leukocytes/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/immunology , Nasopharynx/microbiology , Otitis Media/genetics , Otitis Media/microbiology , Otitis Media/pathology , Phagocytosis , Signal Transduction
13.
Virol J ; 14(1): 227, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29157279

ABSTRACT

BACKGROUND: Postweaning multisystemic wasting syndrome (PMWS) is an emerging disease in swine. Pigs with PMWS are often infected with a variety of other pathogens, including bacteria, viruses and mycoplasm, in addition to porcine circovirus type 2 (PCV2). PCV2 and Haemophilus parasuis serovar 4 (HPS4) coinfection remain epidemic in China. METHODS: Here we report construction of a three-week-old naturally farrowed, colostrum-deprived (NFCD) piglet's infection model and demonstrate that PCV2-infected piglets with the HPS4 coinfection increased the virulence of PCV2 and these pathogens interact acquired PMWS. RESULTS: All the single infected piglets were transiently bacteremic or viremic. All the PCV2/HPS4 coinfected piglets developed PMWS, characterized by dyspnea, anorexia, prostration and lose weight severely. Co-infection with PCV2 and HPS4 resulted in an increased amount of virus in serum and tissues, presented a slower generation and lower levels of antibodies against PCV2. Co-infection with PCV2 and HPS4 resulted in further reductions in total and differential peripheral blood leukocyte counts. Meantime, PCV2/ HPS4 coinfection potentiated the severity of lung and lymphoid lesions by PCV2-associated, increased the virulence of PCV2-antigen and enhanced the incidence of PMWS in piglets. CONCLUSION: Co-infection with PCV2 and HPS4 induce the exacerbation of system injuries and enhance the pathogenicity of PCV2 in piglets.


Subject(s)
Circovirus/pathogenicity , Coinfection/veterinary , Haemophilus Infections/veterinary , Haemophilus parasuis/physiology , Porcine Postweaning Multisystemic Wasting Syndrome/microbiology , Porcine Postweaning Multisystemic Wasting Syndrome/virology , Virulence/physiology , Animals , Antibodies, Viral/blood , China , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , DNA, Viral/blood , Haemophilus Infections/pathology , Haemophilus Infections/virology , Leukocyte Count/veterinary , Polymerase Chain Reaction/veterinary , Porcine Postweaning Multisystemic Wasting Syndrome/pathology , Swine
14.
Vet Res ; 48(1): 28, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28472979

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is one of the most significant and economically important infectious diseases affecting swine worldwide and can predispose pigs to secondary bacterial infections caused by, e.g. Haemophilus parasuis. The aim of the presented study was to compare susceptibility of two different types of macrophages which could be in contact with both pathogens during infection with PRRS virus (PRRSV) and in co-infection with H. parasuis. Alveolar macrophages (PAMs) as resident cells provide one of the first lines of defence against microbes invading lung tissue. On the other hand, monocyte derived macrophages (MDMs) represent inflammatory cells accumulating at the site of inflammation. While PAMs were relatively resistant to cytopathogenic effect caused by PRRSV, MDMs were much more sensitive to PRRSV infection. MDMs infected with PRRSV increased expression of pro-apoptotic Bad, Bax and p53 mRNA. Increased mortality of MDMs may be also related to a higher intensity of ROS production after infection with PRRSV. In addition, MDMs (but not PAMs) infected with H. parasuis alone formed multinucleated giant cells (MGC); these cells were not observed in MDMs infected with both pathogens. Higher sensitivity of MDMs to PRRSV infection, which is associated with limited MDMs survival and restriction of MGC formation, could contribute to the development of multifactorial respiratory disease of swine.


Subject(s)
Coinfection/veterinary , Giant Cells/virology , Haemophilus Infections/veterinary , Haemophilus parasuis , Macrophages/virology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus , Animals , Coinfection/metabolism , Coinfection/pathology , Giant Cells/metabolism , Giant Cells/pathology , Haemophilus Infections/complications , Haemophilus Infections/pathology , Haemophilus Infections/virology , Macrophages/metabolism , Macrophages/pathology , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/pathology , Pyrimidines , Reactive Oxygen Species/metabolism , Sulfonamides , Swine
15.
Eur J Clin Microbiol Infect Dis ; 36(1): 11-18, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27677281

ABSTRACT

The principal aim of this review is to present the current knowledge regarding acute otitis media (AOM) with spontaneous tympanic membrane perforation (STMP) and to address the question of whether AOM with STMP is a disease with specific characteristics or a severe case of AOM. PubMed was used to search for all studies published over the past 15 years using the key words "acute otitis media" and "othorrea" or "spontaneous tympanic membrane perforation". More than 250 articles were found, but only those published in English and providing data on aspects related to perforation of infectious origin were considered. Early Streptococcus pneumoniae infection due to invasive pneumococcal strains, in addition to coinfections and biofilm production due mainly to non-typeable Haemophilus influenzae, seem to be precursors of STMP. However, it is unclear why some children have several STMP episodes during the first years of life that resolve without complications in adulthood, whereas other children develop chronic suppurative otitis media. Although specific aetiological agents appear to be associated with an increased risk of AOM with STMP, further studies are needed to determine whether AOM with STMP is a distinct disease with specific aetiological, clinical and prognostic characteristics or a more severe case of AOM than the cases that occur without STMP. Finally, it is important to identify preventive methods that are useful not only in otitis-prone children with uncomplicated AOM, but also in children with recurrent AOM and those who experience several episodes with STMP.


Subject(s)
Otitis Media/complications , Otitis Media/epidemiology , Tympanic Membrane Perforation/epidemiology , Tympanic Membrane Perforation/pathology , Haemophilus Infections/epidemiology , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Humans , Otitis Media/microbiology , Pneumococcal Infections/epidemiology , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Tympanic Membrane Perforation/microbiology
16.
J Infect Dis ; 214(5): 817-24, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27288538

ABSTRACT

Several human-adapted bacterial pathogens use a phasevarion (ie, a phase-variable regulon) to rapidly and reversibly regulate the expression of many genes, which include known virulence factors, yet the influence of phasevarion-mediated regulation in pathogenesis remains poorly understood. Here we examine the impact of the nontypeable Haemophilus influenzae (NTHI) ModA2 phasevarion on pathogenesis and disease severity in a chinchilla model of experimental otitis media. Chinchillas were challenged with NTHI variant populations that were either inoculated ON and remained ON, inoculated OFF and shifted ON, or inoculated OFF and remained OFF, within the middle ear. We show that populations that shift from OFF to ON within the middle ear induce significantly greater disease severity than populations that are unable to shift. These observations support the importance of phasevarion switching in NTHI pathogenesis and the necessity to considered phasevarion regulation when developing methods to treat and prevent infection.


Subject(s)
Antigenic Variation , Antigens, Bacterial/immunology , Haemophilus Infections/microbiology , Haemophilus Infections/pathology , Haemophilus influenzae/pathogenicity , Otitis Media/microbiology , Otitis Media/pathology , Animals , Antigens, Bacterial/genetics , Chinchilla , Cohort Studies , Disease Models, Animal , Severity of Illness Index
17.
J Infect Dis ; 214(4): 625-33, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27330052

ABSTRACT

Macrophages can polarize and differentiate to regulate initiation, development, and cessation of inflammation during pulmonary infection with nontypeable Haemophilus influenzae (NTHi). However, the underlying molecular mechanisms driving macrophage phenotypic differentiation are largely unclear. Our study investigated the role of Shp2, a Src homology 2 domain-containing phosphatase, in the regulation of pulmonary inflammation and bacterial clearance. Shp2 levels were increased upon NTHi stimulation. Selective inhibition of Shp2 in mice led to an attenuated inflammatory response by skewing macrophages toward alternatively activated macrophage (M2) polarization. Upon pulmonary NTHi infection, Shp2(-/-) mice, in which the gene encoding Shp2 in monocytes/macrophages was deleted, showed an impaired inflammatory response and decreased antibacterial ability, compared with wild-type controls. In vitro data demonstrated that Shp2 regulated activated macrophage (M1) gene expression via activation of p65-nuclear factor-κB signaling, independent of p38 and extracellular regulated kinase-mitogen-activated proteins kinase signaling pathways. Taken together, our study indicates that Shp2 is required to orchestrate macrophage function and regulate host innate immunity against pulmonary bacterial infection.


Subject(s)
Haemophilus Infections/immunology , Haemophilus Infections/pathology , Haemophilus influenzae/immunology , Macrophages/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Female , Macrophages/physiology , Mice, Inbred C57BL , Mice, Knockout
18.
Infect Immun ; 84(7): 2022-2030, 2016 07.
Article in English | MEDLINE | ID: mdl-27113355

ABSTRACT

Accumulating evidence suggests a connection between asthma development and colonization with nontypeable Haemophilus influenzae (NTHi). Specifically, nasopharyngeal colonization of human infants with NTHi within 4 weeks of birth is associated with an increased risk of asthma development later in childhood. Monocytes derived from these infants have aberrant inflammatory responses to common upper respiratory bacterial antigens compared to those of cells derived from infants who were not colonized and do not go on to develop asthma symptoms in childhood. In this study, we hypothesized that early-life colonization with NTHi promotes immune system reprogramming and the development of atypical inflammatory responses. To address this hypothesis in a highly controlled model, we tested whether colonization of mice with NTHi on day of life 3 induced or exacerbated juvenile airway disease using an ovalbumin (OVA) allergy model of asthma. We found that animals that were colonized on day of life 3 and subjected to induction of allergy had exacerbated airway disease as juveniles, in which exacerbated airway disease was defined as increased cellular infiltration into the lung, increased amounts of inflammatory cytokines interleukin-5 (IL-5) and IL-13 in lung lavage fluid, decreased regulatory T cell-associated FOXP3 gene expression, and increased mucus production. We also found that colonization with NTHi amplified airway resistance in response to increasing doses of a bronchoconstrictor following OVA immunization and challenge. Together, the murine model provides evidence for early-life immune programming that precedes the development of juvenile airway disease and corroborates observations that have been made in human children.


Subject(s)
Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus influenzae/immunology , Nasal Mucosa/microbiology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/microbiology , Animals , Bacterial Load , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Haemophilus Infections/pathology , Hypersensitivity/immunology , Hypersensitivity/microbiology , Mice , Mucin 5AC/biosynthesis , Mucin 5AC/genetics , Mucus , Reproductive Tract Infections/pathology
19.
Sex Transm Infect ; 92(1): 29-31, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26139207

ABSTRACT

OBJECTIVES: To describe the clinical characteristics and therapeutic outcomes from male patients diagnosed of Haemophilus spp urethritis. METHODS: A chart review of patients who presented to our hospital from January 2013 to December 2014 with symptoms of acute urethritis in which Haemophilus spp was isolated in their urethral samples was performed. RESULTS: Haemophilus spp was isolated in 52 out of 413 urethral samples (12.6%) received in our laboratory from patients with symptoms of acute urethritis during the study period. Seven cases corresponded to Haemophilus influenzae and 45 cases to Haemophilus parainfluenzae. The most common clinical presentation was mucopurulent urethral discharge (71%). Eight per cent were HIV-infected patients, and 60% were men who have sex with men. Haemophilus spp was isolated as a single pathogen in 6.8% (28 of 413) of cases. Seventeen per cent of Haemophilus spp were ß-lactamase producers. All patients reported having practiced unprotected insertive oral sex the month before consultation, and five of them denied having had another sexual contact apart from this exposure. In all cases in which follow-up was available, empirical treatment achieved a complete clinical resolution. CONCLUSIONS: Haemophilus spp was considered a pathogen in at least 6.8% of the patients from the evaluated area. It affected men regardless their sexual orientation or HIV status. Unprotected oral sex could play a role in its transmission. The limitations of the study (small sample size and lack of a representative control group) do not allow to prove the true pathogenic role of Haemophilus spp in acute urethritis.


Subject(s)
Exudates and Transudates/microbiology , Haemophilus Infections/microbiology , Haemophilus influenzae/isolation & purification , Haemophilus parainfluenzae/isolation & purification , Unsafe Sex/statistics & numerical data , Urethra/pathology , Urethritis/microbiology , Adult , Anti-Bacterial Agents , Bacterial Typing Techniques , Haemophilus Infections/drug therapy , Haemophilus Infections/pathology , Humans , Male , Urethra/microbiology , Urethritis/drug therapy , Urethritis/etiology
20.
J Immunol ; 192(11): 5226-35, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24752444

ABSTRACT

The detrimental impact of tobacco on human health is clearly recognized, and despite aggressive efforts to prevent smoking, close to one billion individuals worldwide continue to smoke. People with chronic obstructive pulmonary disease are susceptible to recurrent respiratory infections with pathogens, including nontypeable Haemophilus influenzae (NTHI), yet the reasons for this increased susceptibility are poorly understood. Because mortality rapidly increases with multiple exacerbations, development of protective immunity is critical to improving patient survival. Acute NTHI infection has been studied in the context of cigarette smoke exposure, but this is the first study, to our knowledge, to investigate chronic infection and the generation of adaptive immune responses to NTHI after chronic smoke exposure. After chronic NTHI infection, mice that had previously been exposed to cigarette smoke developed increased lung inflammation and compromised adaptive immunity relative to air-exposed controls. Importantly, NTHI-specific T cells from mice exposed to cigarette smoke produced lower levels of IFN-γ and IL-4, and B cells produced reduced levels of Abs against outer-membrane lipoprotein P6, with impaired IgG1, IgG2a, and IgA class switching. However, production of IL-17, which is associated with neutrophilic inflammation, was enhanced. Interestingly, cigarette smoke-exposed mice exhibited a similar defect in the generation of adaptive immunity after immunization with P6. Our study has conclusively demonstrated that cigarette smoke exposure has a profound suppressive effect on the generation of adaptive immune responses to NTHI and suggests the mechanism by which prior cigarette smoke exposure predisposes chronic obstructive pulmonary disease patients to recurrent infections, leading to exacerbations and contributing to mortality.


Subject(s)
Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Pneumonia, Bacterial/immunology , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Animals , B-Lymphocytes/immunology , Bacterial Outer Membrane Proteins/pharmacology , Chronic Disease , Female , Haemophilus Infections/pathology , Haemophilus Infections/prevention & control , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Interferon-gamma/immunology , Interleukin-4/immunology , Mice , Pneumonia, Bacterial/chemically induced , Pneumonia, Bacterial/prevention & control , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/prevention & control , Smoking/immunology , Smoking/pathology
SELECTION OF CITATIONS
SEARCH DETAIL