Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 94(2): 512-519, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36725909

RESUMO

BACKGROUND: Escherichia coli and Group B streptococci (GBS) are the main causes of neonatal early-onset sepsis (EOS). Despite antibiotic therapy, EOS is associated with high morbidity and mortality. Dual inhibition of complement C5 and the Toll-like receptor co-factor CD14 has in animal studies been a promising novel therapy for sepsis. METHODS: Whole blood was collected from the umbilical cord after caesarean section (n = 30). Blood was anti-coagulated with lepirudin. C5 inhibitor (eculizumab) and anti-CD14 was added 8 min prior to, or 15 and 30 min after adding E. coli or GBS. Total bacterial incubation time was 120 min (n = 16) and 240 min (n = 14). Cytokines and the terminal complement complex (TCC) were measured using multiplex technology and ELISA. RESULTS: Dual inhibition significantly attenuated TCC formation by 25-79% when adding inhibitors with up to 30 min delay in both E. coli- and GBS-induced inflammation. TNF, IL-6 and IL-8 plasma concentration were significantly reduced by 28-87% in E. coli-induced inflammation when adding inhibitors with up to 30 min delay. The dual inhibition did not significantly reduce TNF, IL-6 and IL-8 plasma concentration in GBS-induced inflammation. CONCLUSION: Dual inhibition of C5 and CD14 holds promise as a potential future treatment for severe neonatal EOS. IMPACT: Neonatal sepsis can cause severe host inflammation with high morbidity and mortality, but there are still no effective adjunctive immunologic interventions available. Adding CD14 and complement C5 inhibitors up to 30 min after incubation of E. coli or Group B streptococci in a human umbilical cord blood model significantly reduced complement activation and cytokine release. Dual inhibition of C5 and CD14 is a potential future therapy to modulate systemic inflammation in severe cases of neonatal sepsis.


Assuntos
Sepse Neonatal , Sepse , Gravidez , Animais , Recém-Nascido , Humanos , Feminino , Complemento C5 , Escherichia coli , Sangue Fetal , Interleucina-6 , Interleucina-8 , Cesárea , Citocinas , Inflamação , Receptores de Lipopolissacarídeos
2.
Tidsskr Nor Laegeforen ; 141(2)2022 02 01.
Artigo em Inglês, Norueguês | MEDLINE | ID: mdl-35107946

RESUMO

We are in the midst of a hectic season for viral respiratory infections. Respiratory syncytial virus is the most common cause of bronchiolitis in children. The guidelines from the Norwegian Society of Pediatricians are clear that oral glucocorticoids have no place in the treatment of bronchiolitis, and antibiotics are rarely needed.


Assuntos
Bronquiolite , Glucocorticoides , Antibacterianos/uso terapêutico , Bronquiolite/tratamento farmacológico , Glucocorticoides/uso terapêutico , Humanos , Lactente
3.
Acta Paediatr ; 110(8): 2316-2325, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33851423

RESUMO

AIM: To systematically review human metabolomic studies investigating metabolic responses in septic neonates. METHODS: A systematic literature search was performed in the databases MEDLINE, EMBASE and Cochrane library up to the 1st of January 2021. We included studies that assessed neonatal sepsis and the following outcomes; (1) change in the metabolism compared to healthy neonates and/or (2) metabolomics compared to traditional diagnostic tools of neonatal sepsis. The screened abstracts were independently considered for eligibility by two researchers. PROSPERO ID: CRD42020164454. RESULTS: The search identified in total 762 articles. Fifteen articles were assessed for eligibility. Four studies were included, with totally 78 neonates. The studies used different diagnostic criteria and had between 1 and 16 sepsis cases. All studies with bacterial sepsis found alterations in the glucose and lactate metabolism, reflecting possible redistribution of glucose consumption from mitochondrial oxidative phosphorylation to the lactate and pentose phosphate pathway. We also found signs of increased oxidative stress and fatty acid oxidation in sepsis cases. CONCLUSION: We found signs of metabolomic signatures in neonatal sepsis. This may lead to better understanding of sepsis pathophysiology and detection of new candidate biomarkers. Results should be validated in large-scale multicentre studies.


Assuntos
Sepse Neonatal , Sepse , Antibacterianos/uso terapêutico , Humanos , Recém-Nascido , Metabolômica , Sepse Neonatal/diagnóstico , Sepse Neonatal/tratamento farmacológico , Projetos de Pesquisa , Sepse/diagnóstico , Sepse/tratamento farmacológico
4.
APMIS ; 127(6): 475-483, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30916807

RESUMO

The global spread of antimicrobial resistance and the increasing number of immune-compromised patients are major challenges in modern medicine. Targeting bacterial virulence or the human host immune system to increase host defence are important strategies in the search for novel antimicrobial drugs. We investigated the inflammatory response of the synthetic short antimicrobial peptide LTX21 in two model systems: a human whole blood ex vivo model and a murine in vivo peritoneum model - both reflecting early innate immune response. In the whole blood model, LTX21 increased the secretion of a range of different cytokines, decreased the level of tumour necrosis factor (TNF) and activated the complement system. In a haemolysis assay, we found 2.5% haemolysis at a LTX21 concentration of 500 mg/L. In the murine model, increased influx of white blood cells (WBCs) and polymorphonuclear neutrophils (PMNs) in the murine peritoneal cavity was observed after treatment with LTX21. In addition, LTX21 increased monocyte chemoattractant protein-1 (MCP-1). In conclusion, LTX21 affected the inflammatory response; the increase in cytokine secretion, complement activation and WBC influx indicates an activated inflammatory response. The present results indicate the impact of LTX21 on the host-pathogen interplay. Whether this will also affect the course of infection has to be investigated.


Assuntos
Anti-Infecciosos/farmacologia , Inflamação/induzido quimicamente , Oligopeptídeos/farmacologia , Animais , Ativação do Complemento/efeitos dos fármacos , Citocinas/biossíntese , Feminino , Hemólise/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C
5.
J Antimicrob Chemother ; 68(9): 2106-10, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23645587

RESUMO

OBJECTIVES: Biofilm-forming Staphylococcus epidermidis is a prevalent cause of peritonitis during peritoneal dialysis. We compared the efficacy of a synthetic antimicrobial peptidomimetic (Ltx21) versus vancomycin in a murine model mimicking a device-related peritonitis. METHODS: Silicone implants, pre-colonized with an S. epidermidis biofilm, were inserted into the peritoneal cavity of BALB/c mice. Three groups (36 mice in each) with pre-colonized implants received intraperitoneal treatment with Ltx21, vancomycin or placebo. Mice were euthanized on day 3 (n = 12), day 6 (n = 12) or day 8 (n = 12) post-implantation. Controls were mice with sterile implants (n = 18) and mice without surgery (n = 6). Bacterial reductions in cfu were analysed from implants and peritoneal fluid (PF). Inflammatory responses in serum and PF were measured. RESULTS: Vancomycin resulted in a stronger reduction in cfu counts, both on pre-colonized implants and in PF, compared with Ltx21 and placebo. Complete bacterial clearance of the implants was not achieved in any of the groups. The implants pre-colonized with S. epidermidis 1457 resulted in a low-grade peritonitis. We observed, only on day 6, a significant increase in the PF leucocyte count in the group with pre-colonized implants compared with the group with sterile implants (P = 0.0364). CONCLUSIONS: Treatment with vancomycin or Ltx21 was not sufficient to achieve complete bacterial clearance of implants, underlining the difficulties of treating such infections. The low-grade infection may attenuate the inflammatory response and contribute to impaired bacterial clearance.


Assuntos
Antibacterianos/administração & dosagem , Infecções Relacionadas a Cateter/tratamento farmacológico , Peptidomiméticos/administração & dosagem , Peritonite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus epidermidis/efeitos dos fármacos , Vancomicina/administração & dosagem , Animais , Carga Bacteriana , Infecções Relacionadas a Cateter/microbiologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Diálise Peritoneal/efeitos adversos , Peritonite/microbiologia , Infecções Estafilocócicas/microbiologia , Resultado do Tratamento
6.
Pediatr Res ; 73(3): 294-300, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23232670

RESUMO

BACKGROUND: Staphylococcus epidermidis (SE) is an important cause of late-onset sepsis in neonates. SE frequently produces a polysaccharide intercellular adhesin (PIA) biofilm, important in the pathogenesis of these infections. Little is known about how the neonatal innate immune system reacts to SE biofilm-associated infections. Our hypothesis was that SE biofilms induce a lower complement activation in neonates as compared with adults. METHODS: Cord blood from term infants (n = 15) and blood from adults (n = 6) were studied in an ex vivo whole-blood sepsis model. A PIA biofilm-producing strain (SE1457) and its isogenic mutant (M10), producing a non-PIA biofilm, were used. RESULTS: Both SE biofilms induced stronger complement activation in adult than in cord blood (P ≤ 0.033). We found lower levels of antibodies toward both PIA (P = 0.002) and the whole bacterium (P = 0.001) in cord vs. adult blood. By contrast, the interleukin-8 (IL-8) and IL-6 secretion were higher in cord than in adult blood (P ≤ 0.002). The PIA biofilm induced stronger complement activation than the non-PIA biofilm. CONCLUSION: We conclude that the neonatal complement system exhibits a maturational deficiency. This may reduce the ability of neonates to combat biofilm-associated SE infections.


Assuntos
Biofilmes , Ativação do Complemento/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus epidermidis/imunologia , Adulto , Ensaio de Imunoadsorção Enzimática , Sangue Fetal/imunologia , Humanos , Recém-Nascido , Interleucina-6/imunologia , Interleucina-8/imunologia , Polissacarídeos Bacterianos/metabolismo , Infecções Estafilocócicas/sangue , Estatísticas não Paramétricas
7.
FEMS Immunol Med Microbiol ; 63(2): 269-80, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22077230

RESUMO

Staphylococcus epidermidis is a frequent cause of nosocomial infections. The central virulence factor of S. epidermidis is biofilm formation. Polysaccharide intercellular adhesin (PIA) constitutes the major biofilm matrix-component. PIA and biofilm have been implicated in S. epidermidis evasion of host immune defence. We examined the effects of S. epidermidis PIA on the inflammatory response with focus on complement activation. We used a human whole-blood ex vivo model of infection and compared the effects of a PIA-positive S. epidermidis strain (SE1457) and its PIA-negative isogenic mutant (M10). The independent effect of purified PIA on complement activation was investigated. In glucose-rich media, the mutant formed a proteinacious DNA-rich biofilm, whereas SE1457 formed a thick PIA-biofilm. In biofilm growth, SE1457 induced a stronger activation of the complement system compared with M10. We verified that purified PIA was independently responsible for a strong activation of the complement system. In contrast, M10 induced higher granulocyte activation by expression of CD11b and higher secretion of cytokines. We conclude that PIA has potent pro-inflammatory properties by activating the complement system. However, in a complex balance of the immune response, the decreased activation of granulocytes and cytokines by a PIA biofilm may limit host eradication of S. epidermidis.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Polissacarídeos Bacterianos/imunologia , Staphylococcus epidermidis/imunologia , Biofilmes/crescimento & desenvolvimento , Sangue/imunologia , Sangue/microbiologia , Citocinas/metabolismo , Deleção de Genes , Granulócitos/imunologia , Experimentação Humana , Humanos , Polissacarídeos Bacterianos/genética , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/fisiologia
8.
Pediatr Res ; 68(3): 237-41, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20703143

RESUMO

The arginine catabolic mobile element (ACME) in Staphylococci encodes several putative virulence factors. ACME appears to have been transferred from Staphylococcus epidermidis into Staphylococcus aureus and is strongly associated with the epidemic and virulent S. aureus USA300. We sought to determine the distribution of ACME in 128 S. epidermidis blood culture isolates from neonates and to assess ACME's impact on antibiotic resistance, biofilm production, invasive capacity, and host inflammatory response. ACME was detected in 15/64 (23%) invasive blood culture isolates and 26/64 (40%) blood culture contaminants (p = 0.02). ACME-positive S. epidermidis isolates displayed less antibiotic resistance (p < 0.001) and were collected from more mature neonates (p = 0.001). Biofilm production was more prevalent among ACME-negative isolates (61/87) compared with ACME positive (18/41; p = 0.004). Among the 64 children considered having an invasive infection, ACME did not influence the maximum C-reactive protein level. In an in vitro whole-blood sepsis model, there were no differences in the inflammatory response between ACME-positive and ACME-negative isolates. We conclude that ACME in S. epidermidis from neonates was associated with less antibiotic resistance and also does not seem to be associated with increased pathogenicity.


Assuntos
Farmacorresistência Bacteriana/genética , Infecções Estafilocócicas/sangue , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/patogenicidade , Fatores de Virulência/genética , Biofilmes/crescimento & desenvolvimento , Proteína C-Reativa/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Recém-Nascido , Staphylococcus epidermidis/imunologia , Estatísticas não Paramétricas
9.
Tidsskr Nor Laegeforen ; 128(23): 2746-9, 2008 Dec 04.
Artigo em Norueguês | MEDLINE | ID: mdl-19079425

RESUMO

BACKGROUND: Coagulase-negative staphylococci (CoNS) may cause serious infections in immunocompromized patients. CoNS often display multiresistance to antibiotics, and biofilm production is the central virulence factor. Our aim was to investigate these factors in CoNS that colonize children with an increased risk of CoNS infections. MATERIAL AND METHOD: We collected CoNS isolates from intravasal catheters (n = 19) and the skin (n = 47) from 30 hospitalized neonates, and CoNS skin isolates from 20 children with cancer before (n = 20) and after (n = 18) six months of cancer treatment. We analyzed antibiotic resistance and biofilm production with phenotypic methods. We used PCR to detect genes that encode antibiotic resistance and biofilm formation. RESULTS: 11 of 19 (58 %) catheter isolates and 14 of 47 (30 %) skin isolates (p = 0.04) produced biofilm. We found an increasing prevalence of oxacillin resistance (20 % versus 67 %, p = 0.004) and gentamicin resistance (15 % versus 67 %, p = 0.003) after six months of cancer treatment. Biofilm positive CoNS isolates displayed higher levels of antibiotic resistance than biofilm-negative isolates. INTERPRETATION: Our results indicate that sick neonates and children hospitalized with cancer are colonized with pathogenic CoNS strains demonstrating virulence- and antibiotic-resistance patterns that are different from those found in CoNS in healthy people who are not hospitalized.


Assuntos
Biofilmes , Infecções Relacionadas a Cateter/microbiologia , Coagulase/metabolismo , Farmacorresistência Bacteriana Múltipla , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus/enzimologia , Infecções Relacionadas a Cateter/tratamento farmacológico , Criança , Farmacorresistência Bacteriana Múltipla/genética , Humanos , Hospedeiro Imunocomprometido , Recém-Nascido , Neoplasias/imunologia , Neoplasias/microbiologia , Fatores de Risco , Pele/microbiologia , Staphylococcus/genética , Staphylococcus/patogenicidade , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...