Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Future Microbiol ; 16: 777-781, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34229445

RESUMO

Aim: To define sensitivity and specificity of Vitek® 2 MICs as phenotypic screening method for carbapenemase-producing Pseudomonas aeruginosa. Materials & methods: We determined Vitek® 2 MICs of antipseudomonal antimicrobials in 130 unrelated carbapenemase-producing P. aeruginosa and 129 carbapenemase-negative P. aeruginosa isolates within a Dutch carbapenemase-surveillance database. We calculated test characteristics of single and combined antimicrobial MICs for carbapenemase production. Results: Vitek® 2 MIC above epidemiological cutoff of both imipenem and tobramycin or ciprofloxacin and tobramycin displayed a sensitivity of 96.2% and specificity of 89.6% for carbapenemase production in P. aeruginosa. Conclusion: Vitek® 2 MIC> epidemiological cut-off values seem sensitive and specific as a phenotypic screening strategy for carbapenemase-producing P. aeruginosa. Combining imipenem and tobramycin or ciprofloxacin and tobramycin performed best as a screening strategy for defining which P. aeruginosa isolates should undergo confirmatory tests for carbapenemase production.


Assuntos
Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa , Proteínas de Bactérias , Ciprofloxacina/farmacologia , Imipenem/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Tobramicina/farmacologia , beta-Lactamases
2.
mSystems ; 6(2)2021 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-33727397

RESUMO

Bacterial microbiota play a critical role in mediating local and systemic immunity, and shifts in these microbial communities have been linked to impaired outcomes in critical illness. Emerging data indicate that other intestinal organisms, including bacteriophages, viruses of eukaryotes, fungi, and protozoa, are closely interlinked with the bacterial microbiota and their host, yet their collective role during antibiotic perturbation and critical illness remains to be elucidated. We employed multi-omics factor analysis (MOFA) to systematically integrate the bacterial (16S rRNA), fungal (intergenic transcribed spacer 1 rRNA), and viral (virus discovery next-generation sequencing) components of the intestinal microbiota of 33 critically ill patients with and without sepsis and 13 healthy volunteers. In addition, we quantified the absolute abundances of bacteria and fungi using 16S and 18S rRNA PCRs and characterized the short-chain fatty acids (SCFAs) butyrate, acetate, and propionate using nuclear magnetic resonance spectroscopy. We observe that a loss of the anaerobic intestinal environment is directly correlated with an overgrowth of aerobic pathobionts and their corresponding bacteriophages as well as an absolute enrichment of opportunistic yeasts capable of causing invasive disease. We also observed a strong depletion of SCFAs in both disease states, which was associated with an increased absolute abundance of fungi with respect to bacteria. Therefore, these findings illustrate the complexity of transkingdom changes following disruption of the intestinal bacterial microbiome.IMPORTANCE While numerous studies have characterized antibiotic-induced disruptions of the bacterial microbiome, few studies describe how these disruptions impact the composition of other kingdoms such as viruses, fungi, and protozoa. To address this knowledge gap, we employed MOFA to systematically integrate viral, fungal, and bacterial sequence data from critically ill patients (with and without sepsis) and healthy volunteers, both prior to and following exposure to broad-spectrum antibiotics. In doing so, we show that modulation of the bacterial component of the microbiome has implications extending beyond this kingdom alone, enabling the overgrowth of potentially invasive fungi and viruses. While numerous preclinical studies have described similar findings in vitro, we confirm these observations in humans using an integrative analytic approach. These findings underscore the potential value of multi-omics data integration tools in interrogating how different components of the microbiota contribute to disease states. In addition, our findings suggest that there is value in further studying potential adjunctive therapies using anaerobic bacteria or SCFAs to reduce fungal expansion after antibiotic exposure, which could ultimately lead to improved outcomes in the intensive care unit (ICU).

3.
PLoS One ; 15(11): e0241748, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33147273

RESUMO

BACKGROUND: An increasing body of evidence is indicating that the gut microbiota modulates pulmonary inflammatory responses. This so-called gut-lung axis might be of importance in a whole spectrum of inflammatory pulmonary diseases such as acute respiratory distress syndrome, chronic obstructive pulmonary disease and pneumonia. Here, we investigate the effect of antibiotic disruption of gut microbiota on immune responses in the lung after a intranasal challenge with lipopolysaccharide (LPS). METHODS/RESULTS: C57Bl/6 mice were treated for two weeks with broad-spectrum antibiotics supplemented to their drinking water. Afterwards, mice and untreated control mice were inoculated intranasally with LPS. Mice were sacrificed 2 and 6 hours post-challenge, after which bronchoalveolar lavage fluid (BALF) and lung tissues were taken. Gut microbiota analysis showed that antibiotic-treated mice had a pronounced reduction in numbers and diversity of bacteria. A modest, but time consistent, significant increase of interleukin (IL)-6 release was seen in BALF of antibiotic treated mice. Release of tumor necrosis factor alpha (TNFα), however, was not statistically different between groups. CONCLUSION: Antibiotic induced microbiota disruption is associated with alterations in host responses during LPS-induced lung inflammation. Further studies are required to determine the clinical relevance of the gut-lung axis in pulmonary infection and inflammation.


Assuntos
Lesão Pulmonar Aguda/etiologia , Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/química , Feminino , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL
4.
Sci Rep ; 10(1): 9918, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32555232

RESUMO

Patients that suffer from sepsis exhibit an early hyper-inflammatory immune response which can lead to organ failure and death. In our study, we assessed the immune modulation in the human in vivo endotoxemia model and compared it to ex vivo LPS stimulation using 38 transcriptomic markers. Blood was collected before and after 4 hours of LPS challenge and tested with the Immune Profiling Panel (IPP) using the FilmArray system. The use of IPP showed that markers from the innate immunity dominated the response to LPS in vivo, mainly markers related to monocytes and neutrophils. Comparing the two models, in vivo and ex vivo, revealed that most of the markers were modulated in a similar pattern (68%). Some cytokine markers such as TNF, IFN-γ and IL-1ß were under-expressed ex vivo compared to in vivo. T-cell markers were either unchanged or up-modulated ex vivo, compared to a down-modulation in vivo. Interestingly, markers related to neutrophils were expressed in opposite directions, which might be due to the presence of cell recruitment and feedback loops in vivo. The IPP tool was able to capture the early immune response in both the human in vivo endotoxemia model, a translational model mimicking the immune response observed in septic patients.


Assuntos
Biomarcadores/sangue , Endotoxemia/sangue , Endotoxemia/imunologia , Imunidade Inata/imunologia , Lipopolissacarídeos/farmacologia , Monócitos/metabolismo , Transcriptoma/efeitos dos fármacos , Adolescente , Adulto , Endotoxemia/induzido quimicamente , Endotoxemia/patologia , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Adulto Jovem
5.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31109950

RESUMO

The Gram-negative intracellular pathogen Burkholderia pseudomallei is the causative agent of melioidosis, an important cause of sepsis in Southeast Asia. Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) is essential for an appropriate immune response during pathogen invasion. In patients with melioidosis, TLR5 is the most abundantly expressed TLR, and a hypofunctional TLR5 variant has been associated with improved survival. Here, we studied the functional role of TLR5 and its ligand flagellin in experimental melioidosis. First, we observed differential TLR5 expression in the pulmonary and hepatic compartments upon infection with B. pseudomallei Next, we found that B. pseudomallei-challenged TLR5-deficient (Tlr5-/- ) mice were more susceptible to infection than wild-type (WT) mice, as demonstrated by higher systemic bacterial loads, increased organ injury, and impaired survival. Lung bacterial loads were not different between the two groups. The phenotype was flagellin independent; no difference in in vivo virulence was observed for the flagellin-lacking mutant MM36 compared to the wild-type B. pseudomallei strain 1026b. Tlr5-/- mice showed a similar impaired antibacterial defense when infected with MM36 or 1026b. Ex vivo experiments showed that TLR5-deficient macrophages display markedly impaired phagocytosis of B. pseudomallei In conclusion, these data suggest that TLR5 deficiency has a detrimental flagellin-independent effect on the host response against pulmonary B. pseudomallei infection.


Assuntos
Melioidose/etiologia , Receptor 5 Toll-Like/fisiologia , Animais , Burkholderia pseudomallei/fisiologia , Feminino , Flagelina/metabolismo , Humanos , Pulmão/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia
6.
J Antimicrob Chemother ; 74(3): 782-786, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30418539

RESUMO

OBJECTIVES: The impact of combination antibiotic therapy on the composition of the intestinal microbiota remains ill-defined. We aimed to assess the effect of a 1 week antibiotic regimen on the intestinal microbiota of healthy humans for a period of up to 31 months. PATIENTS AND METHODS: Thirteen healthy adult men received either no treatment or oral broad-spectrum antibiotics (ciprofloxacin, vancomycin and metronidazole) for 7 days. At four timepoints (prior to treatment, on day 9, day 49 and 8-31 months later) faecal samples were collected and analysed using 16S RNA gene sequencing. RESULTS: The short-term impact of broad-spectrum antibiotics on the gut microbiota was profound, with a loss of diversity and drastic shifts in community composition. In addition, antibiotics significantly reduced the abundance of bacterial taxa with important metabolic functions, such as the production of butyrate. The microbiota showed a remarkable return towards baseline after 8-31 months, but community composition often remained altered from its initial state. CONCLUSIONS: These findings suggest that combined treatment with vancomycin, ciprofloxacin and metronidazole has a profound and long-lasting effect on microbiota composition, the consequences of which remain largely unknown.


Assuntos
Antibacterianos/farmacologia , Ciprofloxacina/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Metronidazol/farmacologia , Vancomicina/farmacologia , Administração Oral , Adolescente , Adulto , Antibacterianos/administração & dosagem , Biodiversidade , Ciprofloxacina/administração & dosagem , Fezes/microbiologia , Voluntários Saudáveis , Humanos , Masculino , Metronidazol/administração & dosagem , Fatores de Tempo , Vancomicina/administração & dosagem , Adulto Jovem
7.
J Immunol ; 201(4): 1241-1252, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29987161

RESUMO

Peptidylarginine deiminase 4 (PAD4) catalyzes citrullination of histones, an important step for neutrophil extracellular trap (NET) formation. We aimed to determine the role of PAD4 during pneumonia. Markers of NET formation were measured in lavage fluid from airways of critically ill patients. NET formation and host defense were studied during pneumonia-derived sepsis caused by Klebsiella pneumoniae in PAD4+/+ and PAD4-/- mice. Patients with pneumosepsis, compared with those with nonpulmonary disease, showed increased citrullinated histone 3 (CitH3) levels in their airways and a trend toward elevated levels of NET markers cell-free DNA and nucleosomes. During murine pneumosepsis, CitH3 levels were increased in the lungs of PAD4+/+ but not of PAD4-/- mice. Combined light and electron microscopy showed NET-like structures surrounding Klebsiella in areas of CitH3 staining in the lung; however, these were also seen in PAD4-/- mice with absent CitH3 lung staining. Moreover, cell-free DNA and nucleosome levels were mostly similar in both groups. Moreover, Klebsiella and LPS could still induce NETosis in PAD4-/- neutrophils. Both groups showed largely similar bacterial growth, lung inflammation, and organ injury. In conclusion, these data argue against a major role for PAD4 in NET formation, host defense, or organ injury during pneumonia-derived sepsis.


Assuntos
Armadilhas Extracelulares/imunologia , Infecções por Klebsiella/imunologia , Desiminases de Arginina em Proteínas/imunologia , Sepse/imunologia , Animais , Armadilhas Extracelulares/enzimologia , Humanos , Infecções por Klebsiella/enzimologia , Klebsiella pneumoniae/imunologia , Camundongos , Camundongos Knockout , Proteína-Arginina Desiminase do Tipo 4 , Sepse/enzimologia
8.
PLoS Negl Trop Dis ; 11(4): e0005548, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28422970

RESUMO

BACKGROUND: Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is an emerging cause of pneumonia-derived sepsis in the tropics. The gut microbiota supports local mucosal immunity and is increasingly recognized as a protective mediator in host defenses against systemic infection. Here, we aimed to characterize the composition and function of the intestinal microbiota during experimental melioidosis. METHODOLOGY/PRINCIPAL FINDINGS: C57BL/6 mice were infected intranasally with B. pseudomallei and sacrificed at different time points to assess bacterial loads and inflammation. In selected experiments, the gut microbiota was disrupted with broad-spectrum antibiotics prior to inoculation. Fecal bacterial composition was analyzed by means of IS-pro, a 16S-23S interspacer region-based profiling method. A marked shift in fecal bacterial composition was seen in all mice during systemic B. pseudomallei infection with a strong increase in Proteobacteria and decrease in Actinobacteria, with an increase in bacterial diversity. We found enhanced early dissemination of B. pseudomallei and systemic inflammation during experimental melioidosis in microbiota-disrupted mice compared with controls. Whole-genome transcriptional profiling of the lung identified several genes that were differentially expressed between mice with a normal or disrupted intestinal microbiota. Genes involved in acute phase signaling, including macrophage-related signaling pathways were significantly elevated in microbiota disrupted mice. Compared with controls, alveolar macrophages derived from antibiotic pretreated mice showed a diminished capacity to phagocytose B. pseudomallei. This might in part explain the observed protective effect of the gut microbiota in the host defense against pneumonia-derived melioidosis. CONCLUSIONS/SIGNIFICANCE: Taken together, these data identify the gut microbiota as a potential modulator of innate immunity during B. pseudomallei infection.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade Inata , Pulmão/imunologia , Melioidose/imunologia , Animais , Antibacterianos/administração & dosagem , Bactérias/genética , Bactérias/imunologia , DNA Espaçador Ribossômico/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fatores Imunológicos/análise , Fatores Imunológicos/genética , Camundongos Endogâmicos C57BL
9.
Virulence ; 8(8): 1683-1694, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28323523

RESUMO

Melioidosis is a severe infectious disease with a high mortality that is endemic in South-East Asia and Northern Australia. The causative pathogen, Burkholderia pseudomallei, is listed as potential bioterror weapon due to its high virulence and potential for easy dissemination. Currently, there is no licensed vaccine for prevention of melioidosis. Here, we explore the use of rapid plasmid DNA vaccination against B. pseudomallei flagellin for protection against respiratory challenge. We tested three flagellin DNA vaccines with different subcellular targeting designs. C57BL/6 mice were vaccinated via skin tattoo on day 0, 3 and 6 before intranasal challenge with B. pseudomallei on day 21. Next, the most effective construct was used as single vaccination on day 0 by tattoo or intranasal formulation. Mice were sacrificed 72 hours post-challenge to assess bacterial loads, cytokine responses, inflammation and microscopic lesions. A construct encoding a cellular secretion signal resulted in the most effective protection against melioidosis via tattooing, with a 10-fold reduction in bacterial loads in lungs and distant organs compared to the empty vector. Strikingly, a single intranasal administration of the same vaccine resulted in >1000-fold lower bacterial loads and increased survival. Pro-inflammatory cytokine responses were significantly diminished and strong reductions in markers for distant organ damage were observed. A rapid vaccination scheme using flagellin DNA tattoo provides significant protection against intranasal challenge with B. pseudomallei, markedly improved by a single administration via airway mucosa. Hence intranasal vaccination with flagellin-encoding DNA may be applicable when acute mass vaccination is indicated and warrants further testing.


Assuntos
Proteínas de Bactérias/administração & dosagem , Burkholderia pseudomallei/imunologia , Flagelina/administração & dosagem , Melioidose/prevenção & controle , Tatuagem/métodos , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Burkholderia pseudomallei/genética , Feminino , Flagelina/genética , Flagelina/imunologia , Humanos , Melioidose/imunologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Vacinas de DNA/genética , Vacinas de DNA/imunologia
11.
Gut ; 66(9): 1623-1630, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27307305

RESUMO

OBJECTIVE: The gut microbiota is essential for the development of the intestinal immune system. Animal models have suggested that the gut microbiota also acts as a major modulator of systemic innate immunity during sepsis. Microbiota disruption by broad-spectrum antibiotics could thus have adverse effects on cellular responsiveness towards invading pathogens. As such, the use of antibiotics may attribute to immunosuppression as seen in sepsis. We aimed to test whether disruption of the gut microbiota affects systemic innate immune responses during endotoxemia in healthy subjects. DESIGN: In this proof-of-principle intervention trial, 16 healthy young men received either no treatment or broad-spectrum antibiotics (ciprofloxacin, vancomycin and metronidazole) for 7 days, after which all were administered lipopolysaccharide intravenously to induce a transient sepsis-like syndrome. At various time points, blood and faeces were sampled. RESULTS: Gut microbiota diversity was significantly lowered by the antibiotic treatment in all subjects. Clinical parameters, neutrophil influx, cytokine production, coagulation activation and endothelial activation during endotoxemia were not different between antibiotic-pretreated and control individuals. Antibiotic treatment had no impact on blood leucocyte responsiveness to various Toll-like receptor ligands and clinically relevant causative agents of sepsis (Streptococcus pneumoniae, Klebsiella pneumoniae, Escherichia coli) during endotoxemia. CONCLUSIONS: These findings suggest that gut microbiota disruption by broad-spectrum antibiotics does not affect systemic innate immune responses in healthy subjects during endotoxemia in humans, disproving our hypothesis. Further research is needed to test this hypothesis in critically ill patients. These data underline the importance of translating findings in mice to humans. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov (NCT02127749; Pre-results).


Assuntos
Ciprofloxacina/farmacologia , Endotoxemia/tratamento farmacológico , Microbioma Gastrointestinal , Imunidade Inata/efeitos dos fármacos , Metronidazol/farmacologia , Sepse , Vancomicina/farmacologia , Adulto , Antibacterianos/farmacologia , Monitoramento de Medicamentos , Endotoxemia/microbiologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Voluntários Saudáveis , Humanos , Imunidade Inata/imunologia , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/fisiologia , Masculino , Sepse/tratamento farmacológico , Sepse/imunologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/fisiologia , Resultado do Tratamento
12.
Intensive Care Med ; 43(1): 59-68, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27837233

RESUMO

PURPOSE: The intestinal microbiota has emerged as a virtual organ with essential functions in human physiology. Antibiotic-induced disruption of the microbiota in critically ill patients may have a negative influence on key energy resources and immunity. We set out to characterize the fecal microbiota composition in critically ill patients both with and without sepsis and to explore the use of microbiota-derived markers for clinical outcome measurements in this setting. METHODS: In this prospective observational cohort study we analyzed the fecal microbiota of 34 patients admitted to the intensive care unit. Fifteen healthy subjects served as controls. The fecal microbiota was phylogenetically characterized by 16S rRNA gene sequencing, and associations with clinical outcome parameters were evaluated. RESULTS: A marked shift in fecal bacterial composition was seen in all septic and non-septic critically ill patients compared with controls, with extreme interindividual differences. In 13 of the 34 patients, a single bacterial genus made up >50% of the gut microbiota; in 4 patients this was even >75%. A significant decrease in bacterial diversity was observed in half of the patients. No associations were found between microbiota diversity, Firmicutes/Bacteroidetes ratio, or Gram-positive/Gram-negative ratio and outcome measurements such as complications and survival. CONCLUSIONS: We observed highly heterogeneous patterns of intestinal microbiota in both septic and non-septic critically ill patients. Nevertheless, some general patterns were observed, including disappearance of bacterial genera with important functions in host metabolism. More detailed knowledge of the short- and long-term health consequences of these major shifts in intestinal bacterial communities is needed.


Assuntos
Antibacterianos/efeitos adversos , Biomarcadores/análise , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Sepse/microbiologia , Idoso , Idoso de 80 Anos ou mais , Estado Terminal , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Países Baixos , Projetos Piloto , Estudos Prospectivos
13.
14.
Clin Transl Gastroenterol ; 7(8): e186, 2016 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-27489950

RESUMO

OBJECTIVES: Broad-spectrum antibiotics disrupt the intestinal microbiota. The microbiota is essential for physiological processes, such as the development of the gut immune system. Recent murine data suggest that the intestinal microbiota also modulates systemic innate immune responses; however, evidence in humans is lacking. METHODS: Twelve healthy young men were given oral broad-spectrum antibiotics (ciprofloxacin 500 mg bid, vancomycin 500 mg tid and metronidazole 500 mg tid) for 7 days. At baseline, 1 day, and 6 weeks after antibiotics, blood and feces were sampled. Whole blood and isolated mononuclear cells were stimulated with selected Toll-like receptor agonists and heat-killed bacteria. Microbiota diversity and composition was determined using bacterial 16S rDNA sequencing. RESULTS: One day after the antibiotic course, microbial diversity was significantly lower compared with baseline. After antibiotic therapy, systemic mononuclear cells produced lower levels of tumor necrosis factor (TNF)-α after ex vivo stimulation with lipopolysaccharide (LPS). This diminished capacity to produce TNF-α was restored 6 weeks after cessation of antibiotic therapy. In whole blood, a reduced capacity to release interleukin (IL)-1ß and IL-6 was observed after LPS stimulation. Antibiotic treatment did not impact on differential leukocyte counts, phagocytosis, and cell surface markers of neutrophils and monocytes. CONCLUSIONS: In this proof-of-principle study of healthy subjects, microbiota disruption by broad-spectrum antibiotics is reversibly associated with decreased systemic cellular responsiveness towards LPS. The implications of these findings in a clinical setting remain to be determined.

15.
PLoS Negl Trop Dis ; 10(6): e0004747, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27253382

RESUMO

BACKGROUND: Triggering receptor expressed on myeloid cells (TREM) -1 and TREM-2 are key regulators of the inflammatory response that are involved in the clearance of invading pathogens. Melioidosis, caused by the "Tier 1" biothreat agent Burkholderia pseudomallei, is a common form of community-acquired sepsis in Southeast-Asia. TREM-1 has been suggested as a biomarker for sepsis and melioidosis. We aimed to characterize the expression and function of TREM-1 and TREM-2 in melioidosis. METHODOLOGY/PRINCIPAL FINDINGS: Wild-type, TREM-1/3 (Trem-1/3-/-) and TREM-2 (Trem-2-/-) deficient mice were intranasally infected with live B. pseudomallei and killed after 24, and/or 72 h for the harvesting of lungs, liver, spleen, and blood. Additionally, survival studies were performed. Cellular functions were further analyzed by stimulation and/or infection of isolated cells. TREM-1 and TREM-2 expression was increased both in the lung and liver of B. pseudomallei-infected mice. Strikingly, Trem-2-/-, but not Trem-1/3-/-, mice displayed a markedly improved host defense as reflected by a strong survival advantage together with decreased bacterial loads, less inflammation and reduced organ injury. Cellular responsiveness of TREM-2, but not TREM-1, deficient blood and bone-marrow derived macrophages (BMDM) was diminished upon exposure to B. pseudomallei. Phagocytosis and intracellular killing of B. pseudomallei by BMDM and alveolar macrophages were TREM-1 and TREM-2-independent. CONCLUSIONS/SIGNIFICANCE: We found that TREM-2, and to a lesser extent TREM-1, plays a remarkable detrimental role in the host defense against a clinically relevant Gram-negative pathogen in mice: TREM-2 deficiency restricts the inflammatory response, thereby decreasing organ damage and mortality.


Assuntos
Regulação da Expressão Gênica/imunologia , Melioidose/imunologia , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Animais , Burkholderia pseudomallei , Citocinas/metabolismo , Inflamação/metabolismo , Pneumopatias/imunologia , Pneumopatias/microbiologia , Pneumopatias/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Receptores Imunológicos/genética , Organismos Livres de Patógenos Específicos , Receptor Gatilho 1 Expresso em Células Mieloides
16.
Shock ; 46(5): 566-574, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27219859

RESUMO

BACKGROUND: Melioidosis, caused by the gram-negative bacterium Burkholderia pseudomallei, is a common cause of community-acquired sepsis in Southeast Asia and Northern Australia. The NLRP3 inflammasome and its downstream product interleukin-1 beta (IL-1ß) have been proposed to play crucial roles in melioidosis. In this study, we characterized the role of IL-1ß more closely and we assessed its therapeutic potential. METHODS: mRNA expression of inflammasome components was determined in isolated leukocytes of 32 healthy controls and 34 patients with sepsis caused by B pseudomallei.Wild-type (WT), NLRP3-deficient (Nlrp3), and Asc mice were infected with B pseudomallei. In additional experiments, infected WT mice were treated with an anti-IL-1ß antibody. After 24, 48, and 72 hours (h) mice were sacrificed and organs were harvested. Furthermore, survival studies were performed. RESULTS: Patients with melioidosis exhibited lower mRNA levels of caspase-1, NLRP3, and ASC. Bacterial dissemination and organ damage were increased in B pseudomallei-infected Nlrp3 and Asc mice, together with a reduced pulmonary cell influx. Anti-IL-1ß treatment of B pseudomallei challenged mice resulted in strongly reduced bacterial counts, organ damage, and pulmonary granulocyte influx together with reduced mortality. Postponement of anti-IL-1ß treatment for 24 h postinfection still protected mice during melioidosis. CONCLUSION: Expression of caspase-1, NLRP3, and ASC is altered in melioidosis patients. In mice, both NLRP3 and ASC contribute to the host defense against melioidosis. Anti-IL-1ß treatment protects mice against B pseudomallei infection and might be a novel treatment strategy in melioidosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Interleucina-1beta/antagonistas & inibidores , Melioidose/tratamento farmacológico , Melioidose/microbiologia , Adolescente , Adulto , Idoso , Animais , Burkholderia pseudomallei/patogenicidade , Modelos Animais de Doenças , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Pessoa de Meia-Idade , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA Mensageiro/genética , Adulto Jovem
17.
Gut ; 65(4): 575-83, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26511795

RESUMO

OBJECTIVE: Pneumonia accounts for more deaths than any other infectious disease worldwide. The intestinal microbiota supports local mucosal immunity and is increasingly recognised as an important modulator of the systemic immune system. The precise role of the gut microbiota in bacterial pneumonia, however, is unknown. Here, we investigate the function of the gut microbiota in the host defence against Streptococcus pneumoniae infections. DESIGN: We depleted the gut microbiota in C57BL/6 mice and subsequently infected them intranasally with S. pneumoniae. We then performed survival and faecal microbiota transplantation (FMT) experiments and measured parameters of inflammation and alveolar macrophage whole-genome responses. RESULTS: We found that the gut microbiota protects the host during pneumococcal pneumonia, as reflected by increased bacterial dissemination, inflammation, organ damage and mortality in microbiota-depleted mice compared with controls. FMT in gut microbiota-depleted mice led to a normalisation of pulmonary bacterial counts and tumour necrosis factor-α and interleukin-10 levels 6 h after pneumococcal infection. Whole-genome mapping of alveolar macrophages showed upregulation of metabolic pathways in the absence of a healthy gut microbiota. This upregulation correlated with an altered cellular responsiveness, reflected by a reduced responsiveness to lipopolysaccharide and lipoteichoic acid. Compared with controls, alveolar macrophages derived from gut microbiota-depleted mice showed a diminished capacity to phagocytose S. pneumoniae. CONCLUSIONS: This study identifies the intestinal microbiota as a protective mediator during pneumococcal pneumonia. The gut microbiota enhances primary alveolar macrophage function. Novel therapeutic strategies could exploit the gut-lung axis in bacterial infections.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas , Pneumonia Pneumocócica/imunologia , Animais , Antibacterianos/farmacologia , Carga Bacteriana , Fezes/microbiologia , Interleucina-10/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Sepse/imunologia , Sepse/metabolismo , Sepse/microbiologia , Streptococcus pneumoniae/imunologia , Transcriptoma , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
18.
Ned Tijdschr Geneeskd ; 157: A5901, 2014.
Artigo em Holandês | MEDLINE | ID: mdl-24780568

RESUMO

The human gut microbiota, formerly known as 'gut flora', may be regarded as an external organ with many physiological functions in metabolism, development of the immune system and defense against pathogens. The adult gut microbiota consist of 1013-1014 micro-organisms. The aggregate genome of these, known as the microbiome, is 100 times larger than the human genome. The gut microbiotica may be involved in the pathogenesis of a range of syndromes, such as inflammatory bowel disease, obesity, diabetes mellitus and atopic disorders. It should be noted that until now most of the studies conducted have been association studies, without proof of causality. This increasing insight has led to identification of new therapeutic strategies, which are currently being investigated in clinical studies. Although the implications of this knowledge for individual patients have yet to become clear, various interventions are conceivable, such as supplementation of nutritional elements, prebiotics or probiotics and feces transplantation.


Assuntos
Trato Gastrointestinal/microbiologia , Microbiota/imunologia , Microbiota/fisiologia , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Obesidade/imunologia , Obesidade/microbiologia , Prebióticos , Probióticos/administração & dosagem , Probióticos/uso terapêutico
19.
Life Sci ; 86(7-8): 225-33, 2010 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19958782

RESUMO

Cathepsin L, a cysteine protease, is considered to be a potential therapeutic target in cancer treatment. Proteases are involved in the development and progression of cancer. Inhibition of activity of specific proteases may slow down cancer progression. In this review, we evaluate recent studies on the inhibition of cathepsin L in cancer. The effects of cathepsin L inhibition as a monotherapy on apoptosis and angiogenesis in cancer are ambiguous. Cathepsin L inhibition seems to reduce invasion and metastasis, but there is concern that selective cathepsin L inhibition induces compensatory activity by other cathepsins. The combination of cathepsin L inhibition with conventional chemotherapy seems to be more promising and has yielded more consistent results. Future research should be focused on the mechanisms and effects of this combination therapy.


Assuntos
Antineoplásicos/uso terapêutico , Catepsina L/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Catepsina L/metabolismo , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA