Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
APMIS ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007242

ABSTRACT

Infective endocarditis (IE) is a severe infection of the inner heart. Even with current standard treatment, the mean in-hospital mortality is as high as 15-20%, and 1-year mortality is up to 40% for left-sided IE. Importantly, IE mortality rates have not changed substantially over the past 30 years, and the incidence of IE is rising. The treatment is challenging due to the bacterial biofilm mode of growth inside the heart valve vegetations, resulting in antibiotic tolerance. Achieving sufficient antibiotic anti-biofilm concentrations in the biofilms of the heart valve vegetations is problematic, even with high-dose and long-term antibiotic therapy. The increasing prevalence of IE caused by antibiotic-resistant bacteria adds to the challenge. Therefore, adjunctive antibiotic-potentiating drug candidates and strategies are increasingly being investigated. Bacteriophage therapy is a reemerging antibacterial treatment strategy for difficult-to-treat infections, mainly biofilm-associated and caused by multidrug-resistant bacteria. However, significant knowledge gaps regarding the safety and efficacy of phage therapy impede more widespread implementation in clinical practice. Hopefully, future preclinical and clinical testing will reveal whether it is a viable treatment. The objective of the present review is to assess whether bacteriophage therapy is a realistic treatment for IE.

2.
Clin Infect Dis ; 77(2): 242-251, 2023 07 26.
Article in English | MEDLINE | ID: mdl-36947131

ABSTRACT

BACKGROUND: In the POET (Partial Oral Endocarditis Treatment) trial, oral step-down therapy was noninferior to full-length intravenous antibiotic administration. The aim of the present study was to perform pharmacokinetic/pharmacodynamic analyses for oral treatments of infective endocarditis to assess the probabilities of target attainment (PTAs). METHODS: Plasma concentrations of oral antibiotics were measured at day 1 and 5. Minimal inhibitory concentrations (MICs) were determined for the bacteria causing infective endocarditis (streptococci, staphylococci, or enterococci). Pharmacokinetic/pharmacodynamic targets were predefined according to literature using time above MIC or the ratio of area under the curve to MIC. Population pharmacokinetic modeling and pharmacokinetic/pharmacodynamic analyses were done for amoxicillin, dicloxacillin, linezolid, moxifloxacin, and rifampicin, and PTAs were calculated. RESULTS: A total of 236 patients participated in this POET substudy. For amoxicillin and linezolid, the PTAs were 88%-100%. For moxifloxacin and rifampicin, the PTAs were 71%-100%. Using a clinical breakpoint for staphylococci, the PTAs for dicloxacillin were 9%-17%.Seventy-four patients at day 1 and 65 patients at day 5 had available pharmacokinetic and MIC data for 2 oral antibiotics. Of those, 13 patients at day 1 and 14 patients at day 5 did only reach the target for 1 antibiotic. One patient did not reach target for any of the 2 antibiotics. CONCLUSIONS: For the individual orally administered antibiotic, the majority reached the target level. Patients with sub-target levels were compensated by the administration of 2 different antibiotics. The findings support the efficacy of oral step-down antibiotic treatment in patients with infective endocarditis.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Humans , Rifampin/therapeutic use , Dicloxacillin/therapeutic use , Linezolid/therapeutic use , Moxifloxacin/therapeutic use , Anti-Bacterial Agents/pharmacology , Endocarditis/drug therapy , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/microbiology , Amoxicillin , Microbial Sensitivity Tests
3.
APMIS ; 130(10): 612-617, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35836366

ABSTRACT

In March 2022, we observed samples with a negative fluorescent signal (60.5%, n = 43) for the influenza A matrix gene and a stronger positive signal for subtype A(H3N2). Forty-three samples were positive in InfA (H3N2) (mean Cq 30.9, range 23.9-35.1), and 26 of the 43 samples were negative in InfA matrix (mean Cq 28.0, range 23.2-30.6). Our multiplex test is a laboratory-developed four-target, four-color influenza A reverse-transcription PCR assay targeting the matrix gene, subtypes A(H3N2) and A(H1N1)pdm09. Several samples were negative when retested on commercial influenza Point-of-Care assays. As the matrix gene is a stand-alone target in most commercial diagnostic assays, we caution against false-negative subtype A test results.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Genetic Drift , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A virus/genetics , Influenza, Human/diagnosis
4.
APMIS ; 130(8): 515-523, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35460117

ABSTRACT

Infective endocarditis (IE) is a serious infection of the inner surface of heart, resulting from minor lesions in the endocardium. The damage induces a healing reaction, which leads to recruitment of fibrin and immune cells. This sterile healing vegetation can be colonized during temporary bacteremia, inducing IE. We have previously established a novel in vitro IE model using a simulated IE vegetation (IEV) model produced from whole venous blood, on which we achieved stable bacterial colonization after 24 h. The bacteria were organized in biofilm aggregates and displayed increased tolerance toward antibiotics. In this current study, we aimed at further characterizing the time course of biofilm formation and the impact on antibiotic tolerance development. We found that a Staphylococcus aureus reference strain, as well as three clinical IE isolates formed biofilms on the IEV after 6 h. When treatment was initiated immediately after infection, the antibiotic effect was significantly higher than when treatment was started after the biofilm was allowed to mature. We could follow the biofilm development microscopically by visualizing growing bacterial aggregates on the IEV. The findings indicate that mature, antibiotic-tolerant biofilms can be formed in our model already after 6 h, accelerating the screening for optimal treatment strategies for IE.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Staphylococcal Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Biofilms , Endocarditis/drug therapy , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/microbiology , Humans , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus
5.
Front Cell Infect Microbiol ; 12: 805964, 2022.
Article in English | MEDLINE | ID: mdl-35186793

ABSTRACT

Patients with infective endocarditis (IE) form a heterogeneous group by age, co-morbidities and severity ranging from stable patients to patients with life-threatening complications with need for intensive care. A large proportion need surgical intervention. In-hospital mortality is 15-20%. The concept of using hyperbaric oxygen therapy (HBOT) in other severe bacterial infections has been used for many decades supported by various preclinical and clinical studies. However, the availability and capacity of HBOT may be limited for clinical practice and we still lack well-designed studies documenting clinical efficacy. In the present review we highlight the potential beneficial aspects of adjunctive HBOT in patients with IE. Based on the pathogenesis and pathophysiological conditions of IE, we here summarize some of the important mechanisms and effects by HBOT in relation to infection and inflammation in general. In details, we elaborate on the aspects and impact of HBOT in relation to the host response, tissue hypoxia, biofilm, antibiotics and pathogens. Two preclinical (animal) studies have shown beneficial effect of HBOT in IE, but so far, no clinical study has evaluated the feasibility of HBOT in IE. New therapeutic options in IE are much needed and adjunctive HBOT might be a therapeutic option in certain IE patients to decrease morbidity and mortality and improve the long-term outcome of this severe disease.


Subject(s)
Endocarditis, Bacterial , Hyperbaric Oxygenation , Animals , Anti-Bacterial Agents/therapeutic use , Combined Modality Therapy , Humans , Treatment Outcome
6.
Front Cell Dev Biol ; 9: 643335, 2021.
Article in English | MEDLINE | ID: mdl-34222225

ABSTRACT

Infective endocarditis (IE) is a life-threatening infective disease with increasing incidence worldwide. From early on, in the antibiotic era, it was recognized that high-dose and long-term antibiotic therapy was correlated to improved outcome. In addition, for several of the common microbial IE etiologies, the use of combination antibiotic therapy further improves outcome. IE vegetations on affected heart valves from patients and experimental animal models resemble biofilm infections. Besides the recalcitrant nature of IE, the microorganisms often present in an aggregated form, and gradients of bacterial activity in the vegetations can be observed. Even after appropriate antibiotic therapy, such microbial formations can often be identified in surgically removed, infected heart valves. Therefore, persistent or recurrent cases of IE, after apparent initial infection control, can be related to biofilm formation in the heart valve vegetations. On this background, the present review will describe potentially novel non-antibiotic, antimicrobial approaches in IE, with special focus on anti-thrombotic strategies and hyperbaric oxygen therapy targeting the biofilm formation of the infected heart valves caused by Staphylococcus aureus. The format is translational from preclinical models to actual clinical treatment strategies.

7.
Int J Antimicrob Agents ; 57(1): 106213, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33256950

ABSTRACT

Cystic fibrosis (CF) patients are at risk of acquiring chronic Pseudomonas aeruginosa lung infections. The biofilm mode of growth of P. aeruginosa induces tolerance to antibiotics and the host response; accordingly, treatment failure occurs. Supplemental azithromycin has proven beneficial in CF owing to potential immunomodulatory mechanisms. Clinical studies have demonstrated a reduction in exacerbations in CF patients by avian IgY anti-Pseudomonas immunotherapy. We hypothesise that azithromycin pre-treatment could potentiate the observed anti-Pseudomonas effect of IgY opsonisation in vivo. Evaluation of phagocytic cell capacity was performed using in vitro exposure of azithromycin pre-treated human polymorphonuclear neutrophils to IgY opsonised P. aeruginosa PAO3. A murine lung infection model using nasal planktonic P. aeruginosa inoculation and successive evaluation 24 h post-infection was used to determine lung bacteriology and subsequent pulmonary inflammation. Combined azithromycin treatment and IgY opsonisation significantly increased bacterial killing compared with the two single-treated groups and controls. In vivo, significantly increased bacterial pulmonary elimination was revealed by combining azithromycin and IgY. A reduction in the inflammatory markers mobiliser granulocyte colony-stimulating factor (G-CSF), macrophage inflammatory protein 2 (MIP-2) and interleukin 1 beta (IL-1ß) paralleled this effect. Combination of azithromycin and anti-Pseudomonas IgY potentiated the killing and pulmonary elimination of P. aeruginosa in vitro and in vivo. The augmented effect of combinatory treatment with azithromycin and IgY constitutes a potential clinical application for improving anti-Pseudomonas strategies.


Subject(s)
Azithromycin/pharmacology , Immunoglobulins/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/immunology , Animals , Anti-Bacterial Agents/pharmacology , Birds/immunology , Colony Count, Microbial , Cystic Fibrosis/microbiology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Female , Immunoglobulins/immunology , Immunotherapy , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred BALB C , Pseudomonas Infections/microbiology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology
8.
J Cyst Fibros ; 19(6): 996-1003, 2020 11.
Article in English | MEDLINE | ID: mdl-32067957

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa is difficult to eradicate from the lungs of cystic fibrosis (CF) patients due to biofilm formation. Organs and blood are independent pharmacokinetic (PK) compartments. Previously, we showed in vitro biofilms behave as independent compartments impacting the pharmacodynamics. The present study investigated this phenomenon in vivo. METHODS: Seaweed alginate beads with P. aeruginosa resembling biofilms, either freshly produced (D0) or incubated for 5 days (D5) were installed s.c in BALB/c mice. Mice (n = 64) received tobramycin 40 mg/kg s.c. and were sacrificed at 0.5, 3, 6, 8, 16 or 24 h after treatment. Untreated controls (n = 14) were sacrificed, correspondingly. Tobramycin concentrations were determined in serum, muscle tissue, lung tissue and beads. Quantitative bacteriology was determined. RESULTS: The tobramycin peak concentrations in serum was 58.3 (±9.2) mg/L, in lungs 7.1 mg/L (±2.3), muscle tissue 2.8 mg/L (±0.5) all after 0.5 h and in D0 beads 19.8 mg/L (±3.5) and in D5 beads 24.8 mg/L (±4.1) (both 3 h). A 1-log killing of P. aeruginosa in beads was obtained at 8h, after which the bacterial level remained stable at 16 h and even increased in D0 beads at 24 h. Using the established diffusion retardation model the free tobramycin concentration inside the beads showed a delayed buildup of 3 h but remained lower than the MIC throughout the 24 h. CONCLUSIONS: The present in vivo study based on tobramycin exposure supports that biofilms behave as independent pharmacological microcompartments. The study indicates, reducing the biofilm matrix would increase free tobramycin concentrations and improve therapeutic effects.


Subject(s)
Biofilms/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Pseudomonas aeruginosa/drug effects , Tobramycin/pharmacokinetics , Alginates/pharmacology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C
9.
Pathog Dis ; 78(5)2020 07 01.
Article in English | MEDLINE | ID: mdl-31116394

ABSTRACT

The majority of chronic wounds are associated with bacterial biofilms recalcitrant to antibiotics and host responses. Immunomodulatory S100A8/A9 is suppressed in Pseudomonas aeruginosa biofilm infected wounds. We aimed at investigating a possible additive effect between S100A8/A9 and ciprofloxacin against biofilms. MATERIALS/METHODS: Thirty-two mice were injected with alginate-embedded P. aeruginosa following a third-degree burn. The mice were randomized into four groups receiving combination ciprofloxacin and S100A8/A9 or monotherapy ciprofloxacin, S100A8/A9 or a placebo and evaluated by host responses and quantitative bacteriology in wounds. In addition, in vitro checkerboard analysis was performed, with P. aeruginosa and ascending S100A8/A9 and ciprofloxacin concentrations. RESULTS: S100A8/A9 augmented the effect of ciprofloxacin in vivo by lowering the bacterial quantity compared to the placebo arm and the two monointervention groups (P < 0.0001). S100A8 and 100A9 were increased in the double-treated group as compared to the monointervention groups (P = 0.032, P = 0.0023). Tissue inhibitor of metalloproteinases-1 and keratinocyte\chemokine chemoattractant-1 were increased in the double-intervention group compared to the S100A8/A9 group (P = 0.050, P = 0.050). No in vitro synergism was detected. CONCLUSION: The observed ciprofloxacin-augmenting effect of S100A8/A9 in vivo was not confirmed by checkerboard analysis, indicating dependence on host cells for the S100A8/A9 effect. S100A8/A9 and ciprofloxacin is a promising therapy for optimizing chronic wound treatment.


Subject(s)
Biofilms/drug effects , Calgranulin A/physiology , Ciprofloxacin/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Wound Infection/immunology , Wound Infection/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Chronic Disease , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Host-Pathogen Interactions , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology
10.
APMIS ; 127(5): 361-371, 2019 May.
Article in English | MEDLINE | ID: mdl-30983040

ABSTRACT

The discovery of antibiotic drugs is considered one of the previous century's most important medical discoveries (Medicine's 10 greatest discoveries. New Haven, CT: Yale University Press, 1998: 263). Appropriate use of antibiotics saves millions of lives each year and prevents infectious complications for numerous people. Still, infections kill unacceptable many people around the world, even in developed countries with easy access to most antibiotic drugs. Optimal use of antibiotics is dependent on the identification of primary and secondary focus, and knowledge on which pathogens to expect in a specific infectious syndrome and information on general patterns of regional antibiotic resistance. Furthermore, sampling for microbiological analysis, knowledge of patient immune status and organ functions, travel history, pharmacokinetics and -dynamics of the different antibiotics and possible biofilm formation are among several factors involved in antibiotic therapy of infectious diseases. The present review aims at describing important considerations when using antibacterial antibiotics and to describe how this is becoming substantially more personalized. The parameters relevant in considering the optimal use of antibiotics to treat infections are shown in Fig. 1 - leading to the most relevant antibiotic therapy for that specific patient. To illustrate this subject, the present review's focus will be on challenges with optimal dosing of antibiotics and risks of underdosing. Especially, in cases highly challenging for achieving the aimed antibiotic effect against bacterial infections - this includes augmented renal clearance (ARC) in sepsis, dosing challenges of antibiotics in pregnancy and against biofilm infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Precision Medicine , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Biofilms , Drug Monitoring , Female , Humans , Kidney/metabolism , Microbial Sensitivity Tests , Pregnancy
11.
Front Immunol ; 9: 2717, 2018.
Article in English | MEDLINE | ID: mdl-30534124

ABSTRACT

Lipocalin-2 is a constituent of the neutrophil secondary granules and is expressed de novo by macrophages and epithelium in response to inflammation. Lipocalin-2 acts in a bacteriostatic fashion by binding iron-loaded siderophores required for bacterial growth. Mycobacterium tuberculosis (M.tb) produces siderophores that can be bound by lipocalin-2. The impact of lipocalin-2 in the innate immune response toward extracellular bacteria has been established whereas the effect on intracellular bacteria, such as M.tb, is less well-described. Here we show that lipocalin-2 surprisingly confers a growth advantage on M.tb in the early stages of infection (3 weeks post-challenge). Using mixed bone marrow chimeras, we demonstrate that lipocalin-2 derived from granulocytes, but not from epithelia and macrophages, leads to increased susceptibility to M.tb infection. In contrast, lipocalin-2 is not observed to promote mycobacterial growth at later stages of M.tb infection. We demonstrate co-localization of granulocytes and mycobacteria within the nascent granulomas at week 3 post-challenge, but not in the consolidated granulomas at week 5. We hypothesize that neutrophil-derived lipocalin-2 acts to supply a source of iron to M.tb in infected macrophages within the immature granuloma, thereby facilitating mycobacterial growth.


Subject(s)
Granulocytes/immunology , Granuloma/immunology , Immunity, Innate , Lipocalin-2/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Granulocytes/pathology , Granuloma/genetics , Granuloma/microbiology , Granuloma/pathology , Lipocalin-2/genetics , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Tuberculosis/genetics , Tuberculosis/pathology
12.
Int J Mol Sci ; 18(7)2017 Jun 26.
Article in English | MEDLINE | ID: mdl-28672877

ABSTRACT

Pseudomonas aeruginosa biofilm maintains and perturbs local host defense, hindering timely wound healing. Previously, we showed that P. aeruginosa suppressed S100A8/A9 of the murine innate host defense. We assessed the potential antimicrobial effect of S100A8/A9 on biofilm-infected wounds in a murine model and P. aeruginosa growth in vitro. Seventy-six mice, inflicted with a full-thickness burn wound were challenged subcutaneously (s.c.) by 106 colony-forming units (CFUs) of P. aeruginosa biofilm. Mice were subsequently randomized into two treatment groups, one group receiving recombinant murine S100A8/A9 and a group of vehicle controls (phosphate-buffered saline, PBS) all treated with s.c. injections daily for up to five days. Wounds were analyzed for quantitative bacteriology and contents of key inflammatory markers. Count of blood polymorphonuclear leukocytes was included. S100A8/A9-treatment ameliorated wound infection, as evaluated by quantitative bacteriology (p ≤ 0.05). In vitro, growth of P. aeruginosa was inhibited dose-dependently by S100A8/A9 in concentrations from 5 to 40 µg/mL, as determined by optical density-measurement (OD-measurement) and quantitative bacteriology. Treatment slightly augmented key inflammatory cytokine Tumor Necrosis Factor-α (TNF-α), but dampened interferon-γ (IFN-γ) levels and blood polymorphonuclear count. In conclusion, topical S100A8/A9 displays remarkable novel immune stimulatory and anti-infective properties in vivo and in vitro. Importantly, treatment by S100A8/A9 provides local infection control. Implications for a role as adjunctive treatment in healing of chronic biofilm-infected wounds are discussed.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biofilms/drug effects , Calgranulin A/administration & dosage , Calgranulin B/administration & dosage , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Wound Infection/immunology , Wound Infection/microbiology , Administration, Topical , Animals , Biomarkers , Chronic Disease , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Immunologic Factors , Inflammation Mediators/metabolism , Leukocyte Count , Mice , Neutrophils/immunology , Neutrophils/metabolism , Pseudomonas Infections/drug therapy , Pseudomonas Infections/metabolism , Wound Infection/drug therapy , Wound Infection/metabolism
13.
Pathog Dis ; 75(7)2017 09 29.
Article in English | MEDLINE | ID: mdl-28645160

ABSTRACT

The impact of Pseudomonas aeruginosa biofilm infections in chronic wounds and clinical implication for healing is receiving increased attention. However, the pathophysiology of host/pathogen interplay is not fully understood. By further revealing the mechanisms, necessary new treatment strategies may be identified. Since the background for chronic wounds is diverse, representative animal models are important. We assessed host response and spontaneous wound closure in the relatively resistant C3H/HeN and the susceptible BALB/c mouse strain. Full-thickness burn wounds were inflicted in 108 mice. Pseudomonas aeruginosa biofilm (106 colony forming units) was injected subcutaneously in 72 mice, euthanised day 4, 7 or 10 days post-infection. Wounds were analysed for neutrophil host response markers: S100A8/A9, keratinocyte-derived chemokine and granulocyte-colony stimulating factor. Total peripheral blood leucocyte and polymorphonuclear count were assessed in parallel. Histopathology evaluated wound inflammatory burden. Photoplanimetry described macroscopical wound closure. Stable chronic wound infection was established in all challenged mice. Pseudomonas aeruginosa biofilm suppressed neutrophil host response in wounds. C3H/HeN mice achieved earlier systemic inflammatory control and healed faster than BALB/c mice. Pseudomonas aeruginosa biofilms perturb host defence thereby inducing a steady state of chronic infection which may impair wound healing. These results indicate therapeutic options for immune modulation of biofilm-infected wounds.


Subject(s)
Biofilms/growth & development , Burns, Electric/microbiology , Calgranulin A/immunology , Calgranulin B/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Wound Infection/microbiology , Animals , Burns, Electric/immunology , Burns, Electric/pathology , Calgranulin A/genetics , Calgranulin B/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Host-Pathogen Interactions , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Neutrophils/immunology , Neutrophils/microbiology , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/physiology , Species Specificity , Wound Healing/immunology , Wound Infection/immunology , Wound Infection/pathology
14.
APMIS ; 125(4): 320-338, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28407429

ABSTRACT

Biofilm infections are one of the modern medical world's greatest challenges. Probably, all non-obligate intracellular bacteria and fungi can establish biofilms. In addition, there are numerous biofilm-related infections, both foreign body-related and non-foreign body-related. Although biofilm infections can present in numerous ways, one common feature is involvement of the host response with significant impact on the course. A special characteristic is the synergy of the innate and the acquired immune responses for the induced pathology. Here, we review the impact of the host response for the course of biofilm infections, with special focus on cystic fibrosis, chronic wounds and infective endocarditis.


Subject(s)
Biofilms , Cystic Fibrosis/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/physiology , Cystic Fibrosis/microbiology , Host-Pathogen Interactions , Humans , Immunity , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics
16.
Echocardiography ; 30(7): E188-91, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23614782

ABSTRACT

A 17-year-old male with a history of newly implanted mechanical valve at the aortic position, presented with fever, rigors, and painful cutaneous abscesses on his lower extremities and was suspected for infective endocarditis. Transthoracic echocardiography (TTE) showed a vegetation-like structure following the movement of the mechanical heart valve (MHV), which eventually proved to be a product of degassing microbubbles (MB).


Subject(s)
Aortic Valve/diagnostic imaging , Aortic Valve/surgery , Artifacts , Diagnostic Errors/prevention & control , Endocarditis/diagnostic imaging , Phospholipids , Staphylococcal Infections/diagnostic imaging , Sulfur Hexafluoride , Adolescent , Contrast Media , Diagnosis, Differential , False Positive Reactions , Heart Valve Prosthesis/adverse effects , Humans , Male , Prosthesis-Related Infections/diagnostic imaging , Prosthesis-Related Infections/etiology , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...