Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 468
1.
Infect Immun ; 92(5): e0045323, 2024 May 07.
Article En | MEDLINE | ID: mdl-38602405

Nontypeable Haemophilus influenzae (NTHi) is a major otitis media (OM) pathogen, with colonization a prerequisite for disease development. Most acute OM is in children <5 years old, with recurrent and chronic OM impacting hearing and learning. Therapies to prevent NTHi colonization and/or disease are needed, especially for young children. Respiratory viruses are implicated in driving the development of bacterial OM in children. We have developed an infant mouse model of influenza-driven NTHi OM, as a preclinical tool for the evaluation of safety and efficacy of clinical therapies to prevent NTHi colonization and the development of OM. In this model, 100% of infant BALB/cARC mice were colonized with NTHi, and all developed NTHi OM. Influenza A virus (IAV) facilitated the establishment of dense (1 × 105 CFU/mL) and long-lasting (6 days) NTHi colonization. IAV was essential for the development of NTHi OM, with 100% of mice in the IAV/NTHi group developing NTHi OM compared with 8% of mice in the NTHi only group. Histological analysis and cytokine measurements revealed that the inflammation observed in the middle ear of the infant mice with OM reflected inflammation observed in children with OM. We have developed the first infant mouse model of NTHi colonization and OM. This ascension model uses influenza-driven establishment of OM and reflects the clinical pathology of bacterial OM developing after a respiratory virus infection. This model provides a valuable tool for testing therapies to prevent or treat NTHi colonization and disease in young children.


Disease Models, Animal , Haemophilus Infections , Haemophilus influenzae , Influenza A virus , Otitis Media , Animals , Otitis Media/microbiology , Haemophilus influenzae/growth & development , Haemophilus influenzae/pathogenicity , Haemophilus influenzae/physiology , Haemophilus Infections/microbiology , Mice , Influenza A virus/pathogenicity , Influenza A virus/growth & development , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/complications , Humans , Animals, Newborn
2.
Microb Pathog ; 190: 106632, 2024 May.
Article En | MEDLINE | ID: mdl-38537762

With the widespread introduction of the Hib conjugate vaccine, Nontypeable Haemophilus influenzae (NTHi) has emerged as the predominant strain globally. NTHi presents a significant challenge as a causative agent of chronic clinical infections due to its high rates of drug resistance and biofilm formation. While current research on NTHi biofilms in children has primarily focused on upper respiratory diseases, investigations into lower respiratory sources remain limited. In this study, we collected 54 clinical strains of lower respiratory tract origin from children. Molecular information and drug resistance features were obtained through whole gene sequencing and the disk diffusion method, respectively. Additionally, an in vitro biofilm model was established. All clinical strains were identified as NTHi and demonstrated the ability to form biofilms in vitro. Based on scanning electron microscopy and crystal violet staining, the strains were categorized into weak and strong biofilm-forming groups. We explored the correlation between biofilm formation ability and drug resistance patterns, as well as clinical characteristics. Stronger biofilm formation was associated with a longer cough duration and a higher proportion of abnormal lung imaging findings. Frequent intake of ß-lactam antibiotics might be associated with strong biofilm formation. While a complementary relationship between biofilm-forming capacity and drug resistance may exist, further comprehensive studies are warranted. This study confirms the in vitro biofilm formation of clinical NTHi strains and establishes correlations with clinical characteristics, offering valuable insights for combating NTHi infections.


Anti-Bacterial Agents , Biofilms , Haemophilus Infections , Haemophilus influenzae , Biofilms/growth & development , Humans , Haemophilus Infections/microbiology , Haemophilus influenzae/physiology , Haemophilus influenzae/isolation & purification , Haemophilus influenzae/genetics , Haemophilus influenzae/drug effects , Haemophilus influenzae/classification , Anti-Bacterial Agents/pharmacology , Child, Preschool , Female , Male , Child , Infant , Microbial Sensitivity Tests , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Microscopy, Electron, Scanning , Drug Resistance, Bacterial , Respiratory System/microbiology , Respiratory System/virology
3.
Front Cell Infect Microbiol ; 12: 969326, 2022.
Article En | MEDLINE | ID: mdl-36081767

Biofilms are multicellular microbial aggregates that can be associated with host mucosal epithelia in the airway, gut, and genitourinary tract. The host environment plays a critical role in the establishment of these microbial communities in both health and disease. These host mucosal microenvironments however are distinct histologically, functionally, and regarding nutrient availability. This review discusses the specific mucosal epithelial microenvironments lining the airway, focusing on: i) biofilms in the human respiratory tract and the unique airway microenvironments that make it exquisitely suited to defend against infection, and ii) how airway pathophysiology and dysfunctional barrier/clearance mechanisms due to genetic mutations, damage, and inflammation contribute to biofilm infections. The host cellular responses to infection that contribute to resolution or exacerbation, and insights about evaluating and therapeutically targeting airway-associated biofilm infections are briefly discussed. Since so many studies have focused on Pseudomonas aeruginosa in the context of cystic fibrosis (CF) or on Haemophilus influenzae in the context of upper and lower respiratory diseases, these bacteria are used as examples. However, there are notable differences in diseased airway microenvironments and the unique pathophysiology specific to the bacterial pathogens themselves.


Cystic Fibrosis , Pseudomonas Infections , Bacteria , Biofilms , Cystic Fibrosis/microbiology , Haemophilus influenzae/physiology , Humans , Pseudomonas aeruginosa/physiology , Respiratory System/pathology
4.
Pediatr Infect Dis J ; 40(9): 792-796, 2021 09 01.
Article En | MEDLINE | ID: mdl-34321442

BACKGROUND: The objective of this study was to determine the prevalence, proportion of encapsulated strains and antibiotic susceptibility of Haemophilus influenzae isolated from young children. METHODS: Children, 6 months to 30 months old, were prospectively enrolled from September 2019 to September 2020 at Rochester, NY, pediatric clinics. H. influenzae isolates from nasopharynx (NP) at healthy visits and disease isolates from NP and middle ear fluid (MEF) at onset of acute otitis media (AOM) were characterized by capsular typing, ß-lactamase production and antibiotic susceptibility. RESULTS: Samples from 565 healthy visits and 130 AOM visits were collected. H. influenzae was detected 5.9% and 27% in the NP from healthy and AOM visits, respectively. In the MEF, H. influenzae was isolated in 43% of samples. Eight percent of H. influenzae isolates were encapsulated, 88% type f. Overall 39.7% of isolates were ß-lactamase producing; 43% for MEF isolates. Ampicillin, trimethoprim/sulfamethoxazole, erythromycin and clarithromycin nonsusceptibility were found in more than 25% of isolates. None of the encapsulated H. influenzae isolates were positive for ß-lactamase production or ampicillin nonsusceptibility. 9.2% of isolates were ß-lactamase negative, ampicillin resistant (ß-lactamase negative, ampicillin resistant + ß-lactamase negative, ampicillin intermediate). CONCLUSIONS: The prevalence of H. influenzae in the NP of young children is very low at times of health, but H. influenzae is highly prevalent in MEF at onset of AOM. Nontypeable H. influenzae accounts for >90% of all H. influenzae isolates. Type f predominated among encapsulated strains. ß-lactamase production and antibiotic nonsusceptibility among H. influenzae strains isolated from the NP and MEF are common.


Bacterial Capsules/physiology , Haemophilus Infections/epidemiology , Haemophilus influenzae/drug effects , Haemophilus influenzae/physiology , Otitis Media/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Child, Preschool , Female , Haemophilus Infections/drug therapy , Haemophilus influenzae/classification , Haemophilus influenzae/genetics , Humans , Infant , Male , Microbial Sensitivity Tests , Otitis Media/epidemiology , Prevalence
5.
PLoS Pathog ; 17(4): e1009513, 2021 04.
Article En | MEDLINE | ID: mdl-33914847

Bacterial meningitis is a major cause of death and disability in children worldwide. Two human restricted respiratory pathogens, Streptococcus pneumoniae and Haemophilus influenzae, are the major causative agents of bacterial meningitis, attributing to 200,000 deaths annually. These pathogens are often part of the nasopharyngeal microflora of healthy carriers. However, what factors elicit them to disseminate and cause invasive diseases, remain unknown. Elevated temperature and fever are hallmarks of inflammation triggered by infections and can act as warning signals to pathogens. Here, we investigate whether these respiratory pathogens can sense environmental temperature to evade host complement-mediated killing. We show that productions of two vital virulence factors and vaccine components, the polysaccharide capsules and factor H binding proteins, are temperature dependent, thus influencing serum/opsonophagocytic killing of the bacteria. We identify and characterise four novel RNA thermosensors in S. pneumoniae and H. influenzae, responsible for capsular biosynthesis and production of factor H binding proteins. Our data suggest that these bacteria might have independently co-evolved thermosensing abilities with different RNA sequences but distinct secondary structures to evade the immune system.


Haemophilus Infections/microbiology , Haemophilus influenzae/immunology , Meningitis, Bacterial/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/immunology , Virulence Factors/metabolism , Bacterial Capsules/metabolism , Base Sequence/genetics , Complement Factor H/metabolism , Environment , Haemophilus influenzae/genetics , Haemophilus influenzae/physiology , Nasopharynx/microbiology , Pneumococcal Infections/genetics , Polysaccharides, Bacterial/metabolism , Streptococcus pneumoniae/physiology , Temperature , Thermosensing
6.
Innate Immun ; 27(3): 251-259, 2021 04.
Article En | MEDLINE | ID: mdl-33646896

Non-typeable Haemophilus influenzae (NTHi) is the most common respiratory pathogen in patients with chronic obstructive disease. Limited data is available investigating the impact of NTHi infections on cellular re-differentiation processes in the bronchial mucosa. The aim of this study was to assess the effects of stimulation with NTHi on the bronchial epithelium regarding cellular re-differentiation processes using primary bronchial epithelial cells harvested from infection-free patients undergoing bronchoscopy. The cells were then cultivated using an air-liquid interface and stimulated with NTHi and TGF-ß. Markers of epithelial and mesenchymal cells were analyzed using immunofluorescence, Western blot and qRT-PCR. Stimulation with both NTHi and TGF-ß led to a marked increase in the expression of the mesenchymal marker vimentin, while E-cadherin as an epithelial marker maintained a stable expression throughout the experiments. Furthermore, expression of collagen 4 and the matrix-metallopeptidases 2 and 9 were increased after stimulation, while the expression of tissue inhibitors of metallopeptidases was not affected by pathogen stimulation. In this study we show a direct pathogen-induced trans-differentiation of primary bronchial epithelial cells resulting in a co-localization of epithelial and mesenchymal markers and an up-regulation of extracellular matrix components.


Bronchi/pathology , Haemophilus Infections/immunology , Haemophilus influenzae/physiology , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Mucosa/physiology , Aged , Cadherins/genetics , Cadherins/metabolism , Cell Transdifferentiation , Cells, Cultured , Collagen Type IV/genetics , Collagen Type IV/metabolism , Female , Humans , Male , Middle Aged , Transforming Growth Factor beta/metabolism , Up-Regulation , Vimentin/genetics , Vimentin/metabolism
7.
Crit Rev Microbiol ; 47(2): 192-205, 2021 Mar.
Article En | MEDLINE | ID: mdl-33455514

Haemophilus influenzae is the most common cause of bacterial infection in the lungs of chronic obstructive pulmonary disease (COPD) patients and contributes to episodes of acute exacerbation which are associated with increased hospitalization and mortality. Due to the ability of H. influenzae to adhere to host epithelial cells, initial colonization of the lower airways can progress to a persistent infection and biofilm formation. This is characterized by changes in bacterial behaviour such as reduced cellular metabolism and the production of an obstructive extracellular matrix (ECM). Herein we discuss the multiple mechanisms by which H. influenzae contributes to the pathogenesis of COPD. In particular, mechanisms that facilitate bacterial adherence to host airway epithelial cells, biofilm formation, and microbial persistence through immune system evasion and antibiotic tolerance will be discussed.


Haemophilus Infections/microbiology , Haemophilus influenzae/growth & development , Pulmonary Disease, Chronic Obstructive/microbiology , Animals , Bacterial Adhesion , Haemophilus influenzae/genetics , Haemophilus influenzae/isolation & purification , Haemophilus influenzae/physiology , Humans , Lung/microbiology
8.
PLoS One ; 16(1): e0243484, 2021.
Article En | MEDLINE | ID: mdl-33411748

Neutrophilic inflammation results in loss of lung function in chronic obstructive pulmonary disease (COPD). Gram-negative bacteria, such as nontypeable Haemophilus influenzae (NTHi), trigger acute exacerbations of COPD (AECOPD) and contribute to chronic lung inflammation. The pro-inflammatory cytokine interleukin-17C (IL-17C) is expressed by airway epithelial cells and regulates neutrophilic chemotaxis. Here, we explored the function of IL-17C in NTHi- and cigarette smoke (CS)-induced models of COPD. Neutrophilic inflammation and tissue destruction were decreased in lungs of IL-17C-deficient mice (Il-17c-/-) chronically exposed to NTHi. Numbers of pulmonary neutrophils were decreased in Il-17c-/- mice after acute exposure to the combination of NTHi and CS. However, Il-17c-/- mice were not protected from CS-induced lung inflammation. In a preliminary patient study, we show that IL-17C is present in sputum samples obtained during AECOPD and associates with disease severity. Concentrations of IL-17C were significantly increased during advanced COPD (GOLD III/IV) compared to moderate COPD (GOLD I/II). Concentrations of IL-17A and IL-17E did not associate with disease severity. Our data suggest that IL-17C promotes harmful pulmonary inflammation triggered by bacteria in COPD.


Disease Progression , Haemophilus influenzae/physiology , Interleukin-17/metabolism , Lung/microbiology , Lung/pathology , Pneumonia/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Sputum/microbiology , Acute Disease , Aged , Animals , Cigarette Smoking/adverse effects , Cytokines/metabolism , Female , Humans , Male , Mice, Inbred C57BL , Neutrophils/pathology
9.
mBio ; 11(6)2020 11 03.
Article En | MEDLINE | ID: mdl-33144377

NTHi is a human-adapted pathogen that colonizes the human respiratory tract. Strains of NTHi express multiple adhesins; however, there is a unique, mutually exclusive relationship between the major adhesins Hia and HMW1 and HMW2 (HMW1/2). Approximately 25% of NTHi strains express Hia, a phase-variable autotransporter protein that has a critical role in colonization of the host nasopharynx. The remaining 75% of strains express HMW1/2. Previous work has shown that the HMW1 and HMW2 proteins mediate binding to 2-3- and 2-6-linked sialic acid glycans found in the human respiratory tract. Here, we show that the high-affinity binding domain of Hia, binding domain 1 (BD1), is responsible for binding to α2-6-sialyllactosamine (2-6 SLN) glycans. BD1 is highly specific for glycans that incorporate the form of sialic acid expressed by humans, N-acetylneuraminic acid (Neu5Ac). We further show that Hia has lower-affinity binding activity for 2-3-linked sialic acid and that this binding activity is mediated via a distinct domain. Thus, Hia with its dual binding activities functionally mimics the combined activities of the HMW1 and HMW2 adhesins. In addition, we show that Hia has a role in biofilm formation by strains of NTHi that express the adhesin. Knowledge of the binding affinity of this major NTHi adhesin and putative vaccine candidate will direct and inform development of future vaccines and therapeutic strategies for this important pathogen.IMPORTANCE Host-adapted bacterial pathogens like NTHi have evolved specific mechanisms to colonize their restricted host niche. Relatively few of the adhesins expressed by NTHi have been characterized as regards their binding affinity at the molecular level. In this work, we show that the major NTHi adhesin Hia preferentially binds to Neu5Ac-α2-6-sialyllactosamine, the form of sialic acid expressed in humans. The receptors targeted by Hia in the human airway mirror those targeted by influenza A virus and indicates the broad importance of sialic acid glycans as receptors for microbes that colonize the human airway.


Adhesins, Bacterial/metabolism , Haemophilus Infections/metabolism , Haemophilus Infections/microbiology , Haemophilus influenzae/physiology , Receptors, Cell Surface/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Adhesins, Bacterial/chemistry , Amino Acid Sequence , Binding Sites , Biofilms , Host-Pathogen Interactions , Humans , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding
10.
mSphere ; 5(3)2020 05 27.
Article En | MEDLINE | ID: mdl-32461275

Nontypeable Haemophilus influenzae (NTHI) colonizes the human nasopharynx, but when the host immune response is dysregulated by upper respiratory tract (URT) virus infection, NTHI can gain access to more distal airway sites and cause disease. The NTHI type IV pilus (T4P) facilitates adherence, benign colonization, and infection, and its majority subunit PilA is in clinical trials as a vaccinogen. To further validate the strategy of immunization with PilA against multiple NTHI-induced diseases, it is important to demonstrate T4P expression under microenvironmental conditions that predispose to NTHI infection of the airway. Because URT infection commonly facilitates NTHI-induced diseases, we examined the influence of ongoing virus infection of respiratory tract epithelial cells on NTHI T4P expression in vitro Polarized primary human airway epithelial cells (HAEs) were sequentially inoculated with one of three common URT viruses, followed by NTHI. Use of a reporter construct revealed that NTHI upregulated pilA promoter activity when cultured with HAEs infected with adenovirus (AV), respiratory syncytial virus (RSV), or rhinovirus (RV) versus that in mock-infected HAEs. Consistent with these results, pilA expression and relative PilA/pilin abundance, as assessed by quantitative reverse transcription-PCR (qRT-PCR) and immunoblot, respectively, were also significantly increased when NTHI was cultured with virus-infected HAEs. Collectively, our data strongly suggest that under conditions of URT virus infection, PilA vaccinogen induction of T4P-directed antibodies is likely to be highly effective against multiple NTHI-induced diseases by interfering with T4P-mediated adherence. We hypothesize that this outcome could thereby limit or prevent the increased load of NTHI in the nasopharynx that characteristically precedes these coinfections.IMPORTANCE Nontypeable Haemophilus influenzae (NTHI) is the predominant bacterial causative agent of many chronic and recurrent diseases of the upper and lower respiratory tracts. NTHI-induced chronic rhinosinusitis, otitis media, and exacerbations of cystic fibrosis and chronic obstructive pulmonary disease often develop during or just after an upper respiratory tract viral infection. We have developed a vaccine candidate immunogen for NTHI-induced diseases that targets the majority subunit (PilA) of the type IV twitching pilus (T4P), which NTHI uses to adhere to respiratory tract epithelial cells and that also plays a role in disease. Here, we showed that NTHI cocultured with virus-infected respiratory tract epithelial cells express significantly more of the vaccine-targeted T4P than NTHI that encounters mock-infected (healthy) cells. These results strongly suggest that a vaccine strategy that targets the NTHI T4P will be effective under the most common predisposing condition: when the human host has a respiratory tract virus infection.


Bacterial Adhesion , Coinfection , Epithelial Cells/microbiology , Epithelial Cells/virology , Fimbriae, Bacterial/genetics , Haemophilus influenzae/genetics , Cells, Cultured , Coinfection/microbiology , Coinfection/virology , Haemophilus influenzae/classification , Haemophilus influenzae/physiology , Humans , Respiratory Syncytial Viruses/pathogenicity , Respiratory System/cytology , Respiratory System/microbiology , Rhinovirus/pathogenicity
11.
Front Immunol ; 11: 485, 2020.
Article En | MEDLINE | ID: mdl-32269571

Inflammatory bowel disease (IBD), which comprises ulcerative colitis and Crohn's disease, is an immune-mediated, chronic-relapsing, disabling disorder which is associated with increased mortality and poor patients' quality of life. Patients with IBD are at increased risk of infections for many reasons. In fact, IBD often requires a lifelong immunosuppressive and/or biologic therapy, both commonly associated with respiratory and opportunistic infections, but also gastrointestinal, urinary tract infections, and sepsis. Moreover, impaired spleen function has been found in a considerable proportion of IBD patients, further increasing the risk of developing infections sustained by encapsulated bacteria, such as S. pneumoniae, H. influenzae, and N. meningitidis. Finally, comorbidities and surgery represent additional risk factors for these patients. Despite the availability of vaccinations against the most common serotypes of encapsulated bacteria, uncertainties still exist regarding a proper vaccination strategy and the actual effectiveness of vaccinations in this particular setting. Aim of this narrative review is to focus on the broad topic of vaccinations against encapsulated bacteria in IBD patients, discussing the clinical impact of infections, predisposing factors, vaccinations strategies, and unmet research and clinical needs.


Bacterial Infections/immunology , Bacterial Vaccines/immunology , Haemophilus influenzae/physiology , Inflammatory Bowel Diseases/immunology , Neisseria meningitidis/physiology , Opportunistic Infections/immunology , Streptococcus pneumoniae/physiology , Animals , Bacterial Infections/etiology , Bacterial Infections/prevention & control , Humans , Inflammatory Bowel Diseases/complications , Opportunistic Infections/etiology , Opportunistic Infections/prevention & control , Vaccination
12.
Int Immunopharmacol ; 83: 106449, 2020 Jun.
Article En | MEDLINE | ID: mdl-32278128

Acute Exacerbation of Chronic Obstructive Pulmonary Disease (AECOPD) is an irreversible inflammatory airways disease responsible for global health burden, involved with a complex condition of immunological change. Exacerbation-mediated neutrophilia is an important factor in the pathogenesis of cigarette smoke-induced AECOPD. Ginsenoside Rg3, a red-ginseng-derived compound, has multiple pharmacological properties such as anti-inflammatory and antitumor activities. Here, we investigated a protective role of Rg3 against AECOPD, focusing on neutrophilia. 14-week-cigarette smoke (CS) exposure and non-typeable Haemophilus inflenzae (NTHi) infection were used to establish the AECOPD murine model. Rg3 (10, 20, 40 mg/kg) was administered intragastrically from the 12th week of CS exposure before infection, and this led to improved lung function and lung morphology, and reduced neutrophilic inflammation, indicating a suppressive effect on neutrophil infiltration by Rg3. Further investigations on the mechanism of Rg3 on neutrophils were carried out using bronchial epithelial cell (BEAS-2B) and neutrophil co-culture and transepithelial migration model. Pre-treatment of neutrophils with Rg3 reduced neutrophil migration, which seemed to be the result of inhibition of phosphatidylinositol (PtdIns) 3-kinases (PI3K) activation within neutrophils. Thus, Rg3 could inhibit exacerbation-induced neutrophilia in COPD by negatively regulating PI3K activities in neutrophils. This study provides a potential natural drug against AECOPD neutrophil inflammation.


Anti-Inflammatory Agents/therapeutic use , Ginsenosides/therapeutic use , Haemophilus Infections/therapy , Haemophilus influenzae/physiology , Lung/pathology , Pulmonary Disease, Chronic Obstructive/therapy , Respiratory Mucosa/metabolism , Animals , Cells, Cultured , Cigarette Smoking/adverse effects , Disease Models, Animal , Disease Progression , Female , Humans , Immune System Diseases , Leukocyte Disorders , Lung/immunology , Mice , Mice, Inbred BALB C , Panax/immunology , Phosphatidylinositol 3-Kinases/metabolism
13.
Cell Rep ; 30(9): 2978-2988.e3, 2020 03 03.
Article En | MEDLINE | ID: mdl-32130901

Bacteria adapt to dynamic changes in the host during chronic and recurrent infections. Bacterial microevolution is one type of adaptation that imparts a selective advantage. We hypothesize that recurrent episodes of disease promote microevolution through genetic mutations that modulate disease severity. We use a pre-clinical model of otitis media (OM) to determine the potential role for microevolution of nontypeable Haemophilus influenzae (NTHI) during sequential episodes of disease. Whole genome sequencing reveals microevolution of hemoglobin binding and lipooligosaccharide (LOS) biosynthesis genes, suggesting that adaptation of these systems is critical for infection. These OM-adapted strains promote increased biofilm formation, inflammation, stromal fibrosis, and an increased propensity to form intracellular bacterial communities (IBCs). Remarkably, IBCs remain for at least one month following clinical resolution of infection, suggesting an intracellular reservoir as a nidus for recurrent OM. Additional approaches for therapeutic design tailored to combat this burdensome disease will arise from these studies.


Disease Progression , Infections/pathology , Acute Disease , Adaptation, Physiological , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Biosynthetic Pathways/genetics , Chinchilla , Fibrosis , Glycosyltransferases/genetics , Haemophilus influenzae/physiology , Haptoglobins/metabolism , Hemoglobins/metabolism , Inflammation/pathology , Lipopolysaccharides/biosynthesis , Otitis Media/genetics , Otitis Media/microbiology , Polymorphism, Single Nucleotide/genetics , Stromal Cells/pathology
14.
BMC Res Notes ; 13(1): 99, 2020 Feb 24.
Article En | MEDLINE | ID: mdl-32093784

OBJECTIVE: For the majority of people with acute sore throat, over-the-counter treatments represent the primary option for symptomatic relief. This study evaluated the in vitro bactericidal activity of lozenges containing the antiseptic hexylresorcinol against five bacteria associated with acute sore throat: Staphylococcus aureus, Streptococcus pyogenes, Moraxella catarrhalis, Haemophilus influenzae and Fusobacterium necrophorum. RESULTS: Hexylresorcinol 2.4 mg lozenges were dissolved into 5 mL of artificial saliva medium. Inoculum cultures were prepared in triplicate for each test organism to give an approximate population of 108 colony-forming units (cfu)/mL. Bactericidal activity was measured by log reduction in cfu. Greater than 3log10 reductions in cfu were observed at 1 min after dissolved hexylresorcinol lozenges were added to S. aureus (log10 reduction cfu/mL ± standard deviation, 3.3 ± 0.2), M. catarrhalis (4.7 ± 0.4), H. influenzae (5.8 ± 0.4) and F. necrophorum (4.5 ± 0.2) and by 5 min for S. pyogenes (4.3 ± 0.4). Hexylresorcinol lozenges achieved a > 99.9% reduction in cfu against all tested organisms within 5 min, which is consistent with the duration for a lozenge to dissolve in the mouth. In conclusion, in vitro data indicate that hexylresorcinol lozenges offer rapid bactericidal activity against organisms implicated in acute sore throat.


Bacterial Infections/drug therapy , Common Cold/drug therapy , Hexylresorcinol/therapeutic use , Oropharynx/drug effects , Administration, Oral , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/therapeutic use , Bacterial Infections/microbiology , Bacterial Load/drug effects , Common Cold/microbiology , Fusobacterium necrophorum/drug effects , Fusobacterium necrophorum/physiology , Haemophilus influenzae/drug effects , Haemophilus influenzae/physiology , Hexylresorcinol/administration & dosage , Humans , Microbial Sensitivity Tests , Moraxella catarrhalis/drug effects , Moraxella catarrhalis/physiology , Oropharynx/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/physiology , Time Factors
15.
Environ Geochem Health ; 42(7): 2137-2145, 2020 Jul.
Article En | MEDLINE | ID: mdl-31845018

Australian Aboriginal populations have unacceptably high rates of bronchiectasis. This disease burden is associated with high rates of detection of pathogenic bacteria, particularly non-typeable Haemophilus influenzae (NTHi). While there is evidence to suggest that exposure to inorganic particulate matter (PM) is associated with worse respiratory infections, no studies have considered the direct effect of this PM on bacterial growth. Nine clinical isolates of pathogenic NTHi were used for this study. Isolates were exposed to two common iron oxides, haematite (Fe2O3) or magnetite (Fe3O4), or quartz (SiO2), as the main constituents of environmental inorganic PM. NTHi isolates were exposed to PM with varying levels of heme to identify whether the response to PM was altered by iron availability. The maximal rate of growth and maximum supported growth were assessed. We observed that inorganic PM was able to modify the maximal growth of selected NTHi isolates. Magnetite and quartz were able to increase maximal growth, while haematite could both increase and suppress the maximal growth. However, these effects varied depending on iron availability and on the bacterial isolate. Our data suggest that inorganic PM may directly alter the growth of pathogenic NTHi. This observation may partly explain the link between exposure to high levels of crustal PM and chronic bacterial infection in Australian Aboriginals.


Haemophilus influenzae/growth & development , Particulate Matter , Australia/epidemiology , Ferric Compounds , Ferrosoferric Oxide , Haemophilus Infections/epidemiology , Haemophilus Infections/microbiology , Haemophilus influenzae/isolation & purification , Haemophilus influenzae/physiology , Humans , Iron/pharmacokinetics , Native Hawaiian or Other Pacific Islander/statistics & numerical data , Silicon Dioxide
16.
Commun Biol ; 2: 340, 2019.
Article En | MEDLINE | ID: mdl-31531401

Multidrug resistance in Gram-negative bacteria can arise by the over-expression of multidrug efflux pumps, which can extrude a wide range of antibiotics. Here we describe the ancestral Haemophilus influenzae efflux pump AcrB (AcrB-Hi). We performed a phylogenetic analysis of hundreds of RND-type transporters. We found that AcrB-Hi is a relatively ancient efflux pump, which nonetheless can export the same range of antibiotics as its evolved colleague from Escherichia coli. AcrB-Hi was not inhibited by the efflux pump inhibitor ABI-PP, and could export bile salts weakly. This points to an environmental adaptation of RND transporters. We also explain the sensitivity of H. influenzae cells to ß-lactams and novobiocin by the outer membrane porin OmpP2. This porin counterbalances the AcrB-Hi efflux by leaking the drugs back into the cells. We hypothesise that multidrug recognition by RND-type pumps is not an evolutionarily acquired ability, and has been present since ancient promiscuous transporters.


Bacterial Proteins/genetics , Haemophilus influenzae/classification , Haemophilus influenzae/physiology , Multidrug Resistance-Associated Proteins/genetics , Phylogeny , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Line , Haemophilus influenzae/drug effects , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Structure-Activity Relationship
18.
BMC Res Notes ; 12(1): 565, 2019 Sep 11.
Article En | MEDLINE | ID: mdl-31506105

OBJECTIVES: Pharyngeal carriers such as H. influenzae seem to constitute the only reservoir and probably the only transmission vehicle of the invasive disease. The aims of this study were to estimate the prevalence of H. influenzae carriage, to characterize antibiotic susceptibility, and to explore genetic diversity of H. influenzae isolates. Sampling was carried out as nasopharynx swabs among children less than 6 years old volunteers. After traditional biochemical tests, isolates were confirmed by targeting omp6 sequence. Following the susceptibility tests, genomic diversity of strains was analyzed by Pulsed-Field Gel Electrophoresis procedure. RESULTS: Out of 328 nasopharynx swabs, 73 strains were identified as H. influenzae. Among H. influenzae isolates, resistance to chloramphenicol (42%) and ampicillin (43%) was observed. Levofloxacin is the most effective antibiotic and the least effect belonged to tetracycline. By genomic analysis of selected H. influenza, 28 PFGE patterns were achieved among which 11 patterns included at least 2 strains. All strains clustered into 25 different clones. The dendrogram analysis of the isolated H. influenzae strains showed that some of these strains had a clonal relationship and common genetic origin. According to our results, antibiotic resistance didn't show any significant correlation with the clonality of strains.


Anti-Bacterial Agents/therapeutic use , Carrier State/drug therapy , Genetic Variation , Haemophilus Infections/drug therapy , Haemophilus influenzae/genetics , Nasopharynx/drug effects , Carrier State/epidemiology , Carrier State/microbiology , Child, Preschool , Cluster Analysis , Electrophoresis, Gel, Pulsed-Field , Female , Haemophilus Infections/epidemiology , Haemophilus Infections/microbiology , Haemophilus influenzae/classification , Haemophilus influenzae/physiology , Humans , Iran/epidemiology , Male , Microbial Sensitivity Tests/methods , Nasopharynx/microbiology , Species Specificity
19.
Crit Rev Microbiol ; 45(5-6): 595-612, 2019.
Article En | MEDLINE | ID: mdl-31502909

Otitis media is a group of inflammatory diseases of the middle ear with great impact on children worldwide. The most common reported bacterial pathogens are Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis. Over the last years, the role of biofilms formed by otopathogens that contribute to otitis media recurrence and chronicity has been established. An improved understanding of the properties of biofilms formed by these bacteria, which factors influence them, and how these affect the host inflammatory response is important for the development of novel strategies for the treatment of otitis media. This review focuses on the biofilm nature that the most prevalent otopathogens adopt in otitis media infections. In addition, new treatment approaches targeting biofilms are highlighted.


Biofilms , Haemophilus influenzae/physiology , Moraxella catarrhalis/physiology , Otitis Media/microbiology , Streptococcus pneumoniae/physiology , Animals , Haemophilus influenzae/genetics , Humans , Moraxella catarrhalis/genetics , Streptococcus pneumoniae/genetics
20.
Infect Immun ; 87(11)2019 11.
Article En | MEDLINE | ID: mdl-31405955

Nontypeable Haemophilus influenzae (NTHi) efficiently colonizes the human nasopharynx asymptomatically but also causes respiratory mucosal infections, including otitis media, sinusitis, and bronchitis. The lipooligosaccharide (LOS) on the cell surface of NTHi displays complex glycans that mimic host structures, allowing it to evade immune recognition. However, LOS glycans are also targets of host adaptive and innate responses. To aid in evasion of these responses, LOS structures exhibit interstrain heterogeneity and are also subject to phase variation, the random on/off switching of gene expression, generating intrastrain population diversity. Specific LOS modifications, including terminal sialylation of the LOS, which exploits host-derived sialic acid (Neu5Ac), can also block recognition of NTHi by bactericidal IgM and complement by mechanisms that are not fully understood. We investigated the LOS sialic acid-mediated resistance of NTHi to antibody-directed killing by serum complement. We identified specific LOS structures extending from heptose III that are targets for binding by naturally occurring bactericidal IgM in serum and are protected by sialylation of the LOS. Phase-variable galactosyltransferases encoded by lic2A and lgtC each add a galactose epitope bound by IgM that results in antibody-dependent killing via the classical pathway of complement. NTHi's survival can be influenced by the expression of phase-variable structures on the LOS that may also depend on environmental conditions, such as the availability of free sialic acid. Identification of surface structures on NTHi representing potential targets for antibody-based therapies as alternatives to antibiotic treatment would thus be valuable for this medically important pathogen.


Complement System Proteins , Haemophilus influenzae/physiology , Immunoglobulin M , Antibodies , Antibodies, Bacterial , Bacterial Proteins , Epitopes , Gene Deletion , Gene Expression Regulation, Bacterial , Humans , Lipopolysaccharides , Magnesium Chloride/pharmacology , N-Acetylneuraminic Acid/pharmacology , Polysaccharides/metabolism , Serum
...