Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1117: 257-279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30980362

RESUMEN

Microbial cells show a strong natural tendency to adhere to surfaces and to colonize them by forming complex communities called biofilms. In this growth mode, biofilm-forming cells encase themselves inside a dense matrix which efficiently protects them against antimicrobial agents and effectors of the immune system. Moreover, at the physiological level, biofilms contain a very heterogeneous cell population including metabolically inactive organisms and persisters, which are highly tolerant to antibiotics. The majority of human infectious diseases are caused by biofilm-forming microorganisms which are responsible for pathologies such as cystic fibrosis, infective endocarditis, pneumonia, wound infections, dental caries, infections of indwelling devices, etc. AMPs are well suited to combat biofilms because of their potent bactericidal activity of broad spectrum (including resting cells and persisters) and their ability to first penetrate and then to disorganize these structures. In addition, AMPs frequently synergize with antimicrobial compounds and were recently reported to repress the molecular pathways leading to biofilm formation. Finally, there is a very active research to develop AMP-containing coatings that can prevent biofilm formation by killing microbial cells on contact or by locally releasing their active principle. In this chapter we will describe these strategies and discuss the perspectives of the use of AMPs as anti-biofilm agents for human therapy and prophylaxis.


Asunto(s)
Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas , Humanos
2.
FEBS J ; 286(8): 1576-1593, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30843356

RESUMEN

Increasing failure of conventional antibiotics to combat bacterial infections requires the urgent development of new antibacterial drugs; a promising class of new drugs based on antimicrobial peptides. Here, we studied the molecular interaction of polycationic synthetic antilipopolysaccharide peptides (SALPs) with various gram-negative and gram-positive bacteria, including resistant strains. The analysis of antimicrobial activity by conventional techniques and atomic force microscopy showed a strict dependence on amino acid (aa) sequences, with the type of amino acid, its position within the primary structure, and the sequence length being critical parameters. By monitoring lipopolysaccharide (LPS)- or bacteria-induced cytokine production in human mononuclear cells and whole blood, we found a direct link between the binding of the lead compound Pep19-2.5 to Salmonella enterica and the anti-inflammatory activity of the peptide. Thermodynamic analysis of Pep19-2.5 binding to the bacterial cell envelope showed an exothermic reaction with saturation characteristics, whereas small-angle X-ray scattering data indicated a direct attachment of Pep19-2.5 to the bacterial cell envelope. This binding preferentially takes place to the LPS outer monolayer, as evidenced by the change in the LPS acyl chain and phosphate vibrational bands seen by Fourier-transform infrared spectroscopy. We report here that the anti-inflammatory activity of Pep19-2.5 is not only connected with neutralization of cell-free bacterial toxins but also with a direct binding of the peptide to the outer leaflet of the bacterial outer membrane.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Antibacterianos/química , Antibacterianos/metabolismo , Calorimetría , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/microbiología , Radioisótopos de Cesio/toxicidad , Citocinas/metabolismo , Citometría de Flujo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Lipopolisacáridos/farmacología , Pruebas de Sensibilidad Microbiana , Microscopía de Fuerza Atómica , Péptidos/síntesis química , Salmonella enterica/efectos de los fármacos , Salmonella enterica/metabolismo , Salmonella enterica/efectos de la radiación , Dispersión del Ángulo Pequeño , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X
3.
Front Immunol ; 9: 1704, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30093904

RESUMEN

The most potent cell wall-derived inflammatory toxins ("pathogenicity factors") of Gram-negative and -positive bacteria are lipopolysaccharides (LPS) (endotoxins) and lipoproteins (LP), respectively. Despite the fact that the former signals via toll-like receptor 4 (TLR4) and the latter via TLR2, the physico-chemistry of these compounds exhibits considerable similarity, an amphiphilic molecule with a polar and charged backbone and a lipid moiety. While the exterior portion of the LPS (i.e., the O-chain) represents the serologically relevant structure, the inner part, the lipid A, is responsible for one of the strongest inflammatory activities known. In the last years, we have demonstrated that antimicrobial peptides from the Pep19-2.5 family, which were designed to bind to LPS and LP, act as anti-inflammatory agents against sepsis and endotoxic shock caused by severe bacterial infections. We also showed that this anti-inflammatory activity requires specific interactions of the peptides with LPS and LP leading to exothermic reactions with saturation characteristics in calorimetry assays. Parallel to this, peptide-mediated neutralization of LPS and LP involves changes in various physical parameters, including both the gel to liquid crystalline phase transition of the acyl chains and the three-dimensional aggregate structures of the toxins. Furthermore, the effectivity of neutralization of pathogenicity factors by peptides was demonstrated in several in vivo models together with the finding that a peptide-based therapy sensitizes bacteria (also antimicrobial resistant) to antibiotics. Finally, a significant step in the understanding of the broad anti-inflammatory function of Pep19-2.5 was the demonstration that this compound is able to block the intracellular endotoxin signaling cascade.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Lipopolisacáridos/efectos adversos , Lipoproteínas/efectos adversos , Péptidos/uso terapéutico , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Endotoxinas/efectos adversos , Endotoxinas/antagonistas & inhibidores , Endotoxinas/química , Humanos , Inflamación/metabolismo , Péptidos/farmacología
4.
Br J Pharmacol ; 175(17): 3581-3593, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29947028

RESUMEN

BACKGROUND AND PURPOSE: Wound healing is a complex process that is essential to provide skin homeostasis. Infection with pathogenic bacteria such as Staphylococcus aureus can lead to chronic wounds, which are challenging to heal. Previously, we demonstrated that the antimicrobial endotoxin-neutralizing peptide Pep19-2.5 promotes artificial wound closure in keratinocytes. Here, we investigated the mechanism of peptide-induced cell migration and if Pep19-2.5 accelerates wound closure in vivo. EXPERIMENTAL APPROACH: Cell migration was examined in HaCaT keratinocytes and P2X7 receptor-overexpressing HEK293 cells using the wound healing scratch assay. The protein expression of phosphorylated ERK1/2, ATP release, calcium influx and mitochondrial ROS were analysed to characterize Pep19-2.5-mediated signalling. For in vivo studies, female BALB/c mice were wounded and infected with methicillin-resistant S. aureus (MRSA) or left non-infected and treated topically with Pep19-2.5 twice daily for 6 days. KEY RESULTS: Specific P2X7 receptor antagonists inhibited Pep19-2.5-induced cell migration and ERK1/2 phosphorylation in keratinocytes and P2X7 receptor-transfected HEK293 cells. ATP release was not increased by Pep19-2.5; however, ATP was required for cell migration. Pep19-2.5 increased cytosolic calcium and mitochondrial ROS, which were involved in peptide-induced migration and ERK1/2 phosphorylation. In both non-infected and MRSA-infected wounds, the wound diameter was reduced already at day 2 post-wounding in the Pep19-2.5-treated groups compared to vehicle, and remained decreased until day 6. CONCLUSIONS AND IMPLICATIONS: Our data suggest the potential application of Pep19-2.5 in the treatment of non-infected and S. aureus-infected wounds and provide insights into the mechanism involved in Pep19-2.5-induced wound healing.


Asunto(s)
Antibacterianos/farmacología , Endotoxinas/antagonistas & inhibidores , Queratinocitos/efectos de los fármacos , Péptidos/farmacología , Agonistas Purinérgicos/farmacología , Receptores Purinérgicos P2X7/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Antibacterianos/química , Línea Celular Transformada , Femenino , Células HEK293 , Humanos , Queratinocitos/citología , Ratones , Ratones Endogámicos BALB C
5.
Exp Mol Med ; 49(6): e345, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28620220

RESUMEN

Sepsis, which is induced by severe bacterial infections, is a major cause of death worldwide, and therapies combating the disease are urgently needed. Because many drugs have failed in clinical trials despite their efficacy in mouse models, the development of reliable animal models of sepsis is in great demand. Several studies have suggested that rabbits reflect sepsis-related symptoms more accurately than mice. In this study, we evaluated a rabbit model of acute sepsis caused by the intravenous inoculation of Salmonella enterica. The model reproduces numerous symptoms characteristic of human sepsis including hyperlactatemia, hyperglycemia, leukopenia, hypothermia and the hyperproduction of several pro-inflammatory cytokines. Hence, it was chosen to investigate the proposed ability of Pep19-2.5-an anti-endotoxic peptide with high affinity to lipopolysaccharide and lipoprotein-to attenuate sepsis-associated pathologies in combination with an antibiotic (ceftriaxone). We demonstrate that a combination of Pep19-2.5 and ceftriaxone administered intravenously to the rabbits (1) kills bacteria and eliminates bacteremia 30 min post challenge; (2) inhibits Toll-like receptor 4 agonists in serum 90 min post challenge; (3) reduces serum levels of pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor α); and (4) reverts to hypothermia and gives rise to temperature values indistinguishable from basal levels 330 min post challenge. The two components of the combination displayed synergism in some of these activities, and Pep19-2.5 notably counteracted the endotoxin-inducing potential of ceftriaxone. Thus, the combination therapy of Pep19-2.5 and ceftriaxone holds promise as a candidate for human sepsis therapy.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Ceftriaxona/uso terapéutico , Péptidos/uso terapéutico , Salmonella enterica/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Ceftriaxona/administración & dosificación , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Células HEK293 , Humanos , Hiperlactatemia , Hipotermia , Interleucina-6/sangre , Leucopenia , Lipopolisacáridos/sangre , Masculino , Péptidos/administración & dosificación , Conejos , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/sangre , Factor de Necrosis Tumoral alfa/sangre
6.
Sci Rep ; 5: 14292, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26390973

RESUMEN

Sepsis, a life-threatening syndrome with increasing incidence worldwide, is triggered by an overwhelming inflammation induced by microbial toxins released into the bloodstream during infection. A well-known sepsis-inducing factor is the membrane constituent of Gram-negative bacteria, lipopolysaccharide (LPS), signalling via Toll-like receptor-4. Although sepsis is caused in more than 50% cases by Gram-positive and mycoplasma cells, the causative compounds are still poorly described. In contradicting investigations lipoproteins/-peptides (LP), lipoteichoic acids (LTA), and peptidoglycans (PGN), were made responsible for eliciting this pathology. Here, we used human mononuclear cells from healthy donors to determine the cytokine-inducing activity of various LPs from different bacterial origin, synthetic and natural, and compared their activity with that of natural LTA and PGN. We demonstrate that LP are the most potent non-LPS pro-inflammatory toxins of the bacterial cell walls, signalling via Toll-like receptor-2, not only in vitro, but also when inoculated into mice: A synthetic LP caused sepsis-related pathological symptoms in a dose-response manner. Additionally, these mice produced pro-inflammatory cytokines characteristic of a septic reaction. Importantly, the recently designed polypeptide Aspidasept(®) which has been proven to efficiently neutralize LPS in vivo, inhibited cytokines induced by the various non-LPS compounds protecting animals from the pro-inflammatory activity of synthetic LP.


Asunto(s)
Antibacterianos/farmacología , Endotoxinas/efectos adversos , Endotoxinas/antagonistas & inhibidores , Lipoproteínas/efectos adversos , Lipoproteínas/antagonistas & inhibidores , Péptidos/farmacología , Sepsis/etiología , Animales , Antibacterianos/síntesis química , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Endotoxemia/tratamiento farmacológico , Endotoxemia/etiología , Endotoxemia/metabolismo , Endotoxemia/mortalidad , Femenino , Bacterias Gramnegativas/inmunología , Células HEK293 , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/química , Lipoproteínas/química , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Péptidos/síntesis química , Peptidoglicano/efectos adversos , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Sepsis/mortalidad , Staphylococcus aureus/inmunología , Ácidos Teicoicos/efectos adversos
7.
PLoS One ; 10(7): e0133291, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26197109

RESUMEN

Sepsis is still a major cause of death and many efforts have been made to improve the physical condition of sepsis patients and to reduce the high mortality rate associated with this disease. While achievements were implemented in the intensive care treatment, all attempts within the field of novel therapeutics have failed. As a consequence new medications and improved patient stratification as well as a thoughtful management of the support therapies are urgently needed. In this study, we investigated the simultaneous administration of ibuprofen as a commonly used nonsteroidal anti-inflammatory drug (NSAID) and Pep19-2.5 (Aspidasept), a newly developed antimicrobial peptide. Here, we show a synergistic therapeutic effect of combined Pep19-2.5-ibuprofen treatment in an endotoxemia mouse model of sepsis. In vivo protection correlates with a reduction in plasma levels of both tumor necrosis factor α and prostaglandin E, as a likely consequence of Pep19-2.5 and ibuprofen-dependent blockade of TLR4 and COX pro-inflammatory cascades, respectively. This finding is further characterised and confirmed in a transcriptome analysis of LPS-stimulated human monocytes. The transcriptome analyses showed that Pep19-2.5 and ibuprofen exerted a synergistic global effect both on the number of regulated genes as well as on associated gene ontology and pathway expression. Overall, ibuprofen potentiated the anti-inflammatory activity of Pep19-2.5 both in vivo and in vitro, suggesting that NSAIDs could be useful to supplement future anti-sepsis therapies.


Asunto(s)
Antiinfecciosos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Ibuprofeno/uso terapéutico , Inflamación/tratamiento farmacológico , Péptidos/uso terapéutico , Sepsis/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Antiinfecciosos/química , Antiinfecciosos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Dinoprostona/inmunología , Sinergismo Farmacológico , Endotoxemia/tratamiento farmacológico , Endotoxemia/inmunología , Femenino , Humanos , Ibuprofeno/farmacología , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Péptidos/química , Péptidos/farmacología , Sepsis/inmunología , Receptor Toll-Like 4/inmunología , Transcriptoma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...