Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Rep ; 9(1): 3947, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850651

RESUMEN

UDP-N-acetylglucosamine (UDP-GlcNAc) acyltransferase (LpxA) catalyzes the first step of lipid A biosynthesis, the transfer of an R-3-hydroxyacyl chain from its acyl carrier protein (ACP) to the 3-OH group of UDP-GlcNAc. Essential in the growth of Gram-negative bacteria, LpxA is a logical target for antibiotics design. A pentadecapeptide (Peptide 920) with high affinity towards LpxA was previously identified in a phage display library. Here we created a small library of systematically designed peptides with the length of four to thirteen amino acids using Peptide 920 as a scaffold. The concentrations of these peptides at which 50% of LpxA is inhibited (IC50) range from 50 nM to >100 µM. We determined the crystal structure of E. coli LpxA in a complex with a potent inhibitor. LpxA-inhibitor interaction, solvent model and all contributing factors to inhibitor efficacy were well resolved. The peptide primarily occludes the ACP binding site of LpxA. Interactions between LpxA and the inhibitor are different from those in the structure of Peptide 920. The inhibitory peptide library and the crystal structure of inhibitor-bound LpxA described here may further assist in the rational design of inhibitors with antimicrobial activity that target LpxA and potentially other acyltransferases.


Asunto(s)
Antibacterianos/farmacología , Diseño de Fármacos , Péptidos/farmacología , Uridina Difosfato N-Acetilglucosamina/antagonistas & inhibidores , Antibacterianos/química , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Concentración 50 Inhibidora , Lípido A/antagonistas & inhibidores , Lípido A/biosíntesis , Biblioteca de Péptidos , Péptidos/química
2.
Artículo en Inglés | MEDLINE | ID: mdl-29938202

RESUMEN

Lipid A is an essential basal component of lipopolysaccharide of most Gram-negative bacteria. Inhibitors targeting LpxC, a conserved enzyme in lipid A biosynthesis, are antibiotic candidates against Gram-negative pathogens. Here we report the characterization of the role of lipid A in Coxiella burnetii growth in axenic media, monkey kidney cells (BGMK and Vero), and macrophage-like THP-1 cells by using a potent LpxC inhibitor -LPC-011. We first determined the susceptibility of C. burnetii LpxC to LPC-011 in a surrogate E. coli model. In E. coli, the minimum inhibitory concentration (MIC) of LPC-011 against C. burnetii LpxC is < 0.05 µg/mL, a value lower than the inhibitor's MIC against E. coli LpxC. Considering the inhibitor's problematic pharmacokinetic properties in vivo and Coxiella's culturing time up to 7 days, the stability of LPC-011 in cell cultures was assessed. We found that regularly changing inhibitor-containing media was required for sustained inhibition of C. burnetii LpxC in cells. Under inhibitor treatment, Coxiella has reduced growth yields in axenic media and during replication in non-phagocytic cells, and has a reduced number of productive vacuoles in such cells. Inhibiting lipid A biosynthesis in C. burnetii by the inhibitor was shown in a phase II strain transformed with chlamydial kdtA. This exogenous KdtA enzyme modifies Coxiella lipid A with an α-Kdo-(2 → 8)-α-Kdo epitope that can be detected by anti-chlamydia genus antibodies. In inhibitor-treated THP-1 cells, Coxiella shows severe growth defects characterized by poor vacuole formation and low growth yields. Coxiella progenies prepared from inhibitor-treated cells retain the capability of normally infecting all tested cells in the absence of the inhibitor, which suggests a dispensable role of lipid A for infection and early vacuole development. In conclusion, our data suggest that lipid A has significance for optimal development of Coxiella-containing vacuoles, and for robust multiplication of C. burnetii in macrophage-like THP-1 cells. Unlike many bacteria, C. burnetii replication in axenic media and non-phagocytic cells was less dependent on normal lipid A biosynthesis.


Asunto(s)
Cultivo Axénico/métodos , Coxiella burnetii/crecimiento & desarrollo , Coxiella burnetii/patogenicidad , Lípido A/antagonistas & inhibidores , Macrófagos/microbiología , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Chlorocebus aethiops , Coxiella burnetii/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Humanos , Ácidos Hidroxámicos/farmacología , Lípido A/genética , Macrófagos/efectos de los fármacos , Células THP-1 , Treonina/análogos & derivados , Treonina/farmacología , Vacuolas/efectos de los fármacos , Vacuolas/microbiología , Células Vero
3.
Microb Pathog ; 94: 2-11, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26318142

RESUMEN

OBJECTIVE: CL(14-25), a dodecapeptide of cyanate lyase from rice, is a novel cationic α-helical antimicrobial peptide. In this study, we examined inhibitory ability of CL(14-25) against endotoxic activities of lipopolysaccharides (LPSs) from Escherichia coli and periodontal pathogenic Aggregatibacter actinomycetemcomitans. METHODS: Endotoxin-neutralizing activity of CL(14-25) was evaluated by inhibition to induction of cytokine and nitric oxide in human aortic endothelial cells (HAECs) and RAW264 mouse macrophage cells, respectively. Protective effect of CL(14-25) was determined in mice against lethal toxicity of LPS. RESULTS: IL-6 in HAECs was induced by stimulation with LPS preparations of A. actinomycetemcomitans and E. coli tested in this study, and addition of CL(14-25) to the medium caused inhibition of their induction in a dose-dependent manner. CL(14-25) inhibited NO induction in RAW264 cells by a smooth type LPS of E. coli O55:B5 and an Rc type LPS of E. coli J5 as well as lipid A of E. coli R515 in a dose-dependent manner. Simultaneous injection of E. coli O55:B5 LPS and CL(14-25) in BALB/c mice resulted in prevention of lethal toxicity of the former. The results of a Limulus amebocyte lysate assay and surface plasmon resonance analysis of interaction between CL(14-25) and E. coli LPS or lipid A showed that CL(14-25) specifically binds to a lipid A moiety of LPS. CONCLUSION: The results of present study suggest that CL(14-25) has a potential to be used as a nutraceutical agent for periodontal therapy.


Asunto(s)
Aggregatibacter actinomycetemcomitans/metabolismo , Liasas de Carbono-Nitrógeno/química , Escherichia coli/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Aggregatibacter actinomycetemcomitans/química , Animales , Citocinas/biosíntesis , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Células Endoteliales/efectos de los fármacos , Escherichia coli/química , Humanos , Interleucina-6/biosíntesis , Lípido A/antagonistas & inhibidores , Lípido A/química , Lípido A/toxicidad , Lipopolisacáridos/química , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Oryza/enzimología , Fragmentos de Péptidos/química , Células RAW 264.7
4.
Innate Immun ; 18(1): 171-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21382909

RESUMEN

Lipid A, the active moiety of LPS, exerts its effects through interaction with TLR4, triggering a signalling cascade that results in the release of pro-inflammatory cytokines. Eritoran is a lipid A analogue that competes with LPS for binding to TLR4; however, after intravenous administration, it undergoes a time-dependent deactivation as a consequence of binding to high-density lipoproteins (HDLs). The site of eritoran association with HDL remains unknown. Therefore the aim of this study was to determine if HDL-associated apolipoproteins A1, A2, serum amyloid A (SAA) and C1, inhibit the ability of eritoran to block LPS-induced TNF-α release from whole blood. Eritoran activity after LPS stimulation in human whole blood was assessed in the presence of reconstituted HDL (rHDL) containing different apos. In rHDL, the major apolipoproteins in both the healthy and septic state, A1 and SAA, caused a significant reduction in eritoran antagonistic activity and had a greater effect than minor apolipoproteins A2 and C1. Apolipoproteins associated with HDL are likely to facilitate eritoran deactivation. Apolipoproteins A1 and SAA should be of particular focus as they are the major apos found on HDL in both the healthy and septic state. Further evaluation of the physical association between apolipoproteins and eritoran should be explored.


Asunto(s)
Células Sanguíneas/efectos de los fármacos , Disacáridos/antagonistas & inhibidores , Lípido A/antagonistas & inhibidores , Lipoproteínas HDL/inmunología , Fosfatos de Azúcar/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/química , Apolipoproteína A-II/metabolismo , Unión Competitiva , Células Sanguíneas/inmunología , Células Sanguíneas/metabolismo , Células Sanguíneas/patología , Disacáridos/farmacología , Humanos , Estructura Molecular , Proteína Amiloide A Sérica/metabolismo , Fosfatos de Azúcar/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Endod ; 35(5): 668-72, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19410080

RESUMEN

INTRODUCTION: It is important to develop an antimicrobial agent without any damage on dental pulp. In the present study, we examined whether pretreatment of bacterial lipopolysaccharides (LPS) with ozonated water (O(3)aq) improves LPS-induced responses of rat odontoblastic cell line, KN-3. METHODS: After the pretreatment of LPS with O(3)aq, effects of LPS and O(3)aq-treated LPS on cell viability; calcification ability; expression of cyclooxygenase 2 (COX-2), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-alpha); and activation of p38 of KN-3 cells were examined. RESULTS: The formation of mineralized nodules by KN-3 cells was suppressed by LPS, whereas that suppression was inhibited by the pretreatment of LPS with ozonated water. We also found that LPS-induced expression of COX-2, IL-6, and TNF-alpha and p38 activation were markedly suppressed when LPS was pretreated with ozonated water. Furthermore, expression of COX-2, IL-6, and TNF-alpha by LPS were mainly induced through p38 activation. CONCLUSION: These results suggest that odontoblastic cells exhibit inflammatory responses against LPS and that ozonated water has the ability to improve LPS-induced inflammatory responses and suppression of odontoblastic properties of KN-3 cells through direct inhibition of LPS.


Asunto(s)
Antiinfecciosos/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Odontoblastos/efectos de los fármacos , Oxidantes Fotoquímicos/farmacología , Ozono/farmacología , Aggregatibacter actinomycetemcomitans , Animales , Butadienos/farmacología , Calcificación Fisiológica/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 1/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Escherichia coli , Imidazoles/farmacología , Interleucina-6/análisis , Lípido A/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Piridinas/farmacología , Ratas , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
6.
Biopolymers ; 91(3): 207-20, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19107926

RESUMEN

The anti-lipopolysaccharide factor ALF-Pm3 is a 98-residue protein identified in hemocytes from the black tiger shrimp Penaeus monodon. It was expressed in Pichia pastoris from the constitutive glyceraldehyde-3-phosphate dehydrogenase promoter as a folded and (15)N uniformly labeled rALF-Pm3 protein. Its 3D structure was established by NMR and consists of three alpha-helices packed against a four-stranded beta-sheet. The C(34)-C(55) disulfide bond was shown to be essential for the structure stability. By using surface plasmon resonance, we demonstrated that rALF-Pm3 binds to LPS, lipid A and to OM-174, a soluble analogue of lipid A. Biophysical studies of rALF-Pm3/LPS and rALF-Pm3/OM-174 complexes indicated rather high molecular sized aggregates, which prevented us to experimentally determine by NMR the binding mode of these lipids to rALF-Pm3. However, on the basis of striking structural similarities to the FhuA/LPS complex, we designed an original model of the possible lipid A-binding site of ALF-Pm3. Such a binding site, located on the ALF-Pm3 beta-sheet and involving seven charged residues, is well conserved in ALF-L from Limulus polyphemus and in ALF-T from Tachypleus tridentatus. In addition, our model is in agreement with experiments showing that beta-hairpin synthetic peptides corresponding to ALF-L beta-sheet bind to LPS. Delineating lipid A-binding site of ALFs will help go further in the de novo design of new antibacterial or LPS-neutralizing drugs.


Asunto(s)
Crustáceos/química , Hormonas de Invertebrados/química , Hormonas de Invertebrados/metabolismo , Lípido A/antagonistas & inhibidores , Lípido A/metabolismo , Modelos Moleculares , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Hormonas de Invertebrados/genética , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Homología Estructural de Proteína , Resonancia por Plasmón de Superficie
7.
Mol Microbiol ; 67(2): 264-77, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18047581

RESUMEN

One-third of the lipid A found in the Escherichia coli outer membrane contains an unsubstituted diphosphate unit at position 1 (lipid A 1-diphosphate). We now report an inner membrane enzyme, LpxT (YeiU), which specifically transfers a phosphate group to lipid A, forming the 1-diphosphate species. (32)P-labelled lipid A obtained from lpxT mutants do not produce lipid A 1-diphosphate. In vitro assays with Kdo(2)-[4'-(32)P]lipid A as the acceptor shows that LpxT uses undecaprenyl pyrophosphate as the substrate donor. Inhibition of lipid A 1-diphosphate formation in wild-type bacteria was demonstrated by sequestering undecaprenyl pyrophosphate with the cyclic polypeptide antibiotic bacitracin, providing evidence that undecaprenyl pyrophosphate serves as the donor substrate within whole bacteria. LpxT-catalysed phosphorylation is dependent upon transport of lipid A across the inner membrane by MsbA, a lipid A flippase, indicating a periplasmic active site. In conclusion, we demonstrate a novel pathway in the periplasmic modification of lipid A that is directly linked to the synthesis of undecaprenyl phosphate, an essential carrier lipid required for the synthesis of various bacterial polymers, such as peptidoglycan.


Asunto(s)
Lípido A/metabolismo , Periplasma/enzimología , Fosfatos de Poliisoprenilo/biosíntesis , Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/farmacología , Bacitracina/farmacología , Proteínas Bacterianas/metabolismo , Escherichia coli K12/enzimología , Escherichia coli K12/genética , Lípido A/antagonistas & inhibidores , Lípidos de la Membrana/metabolismo , Mutación , Peptidil Transferasas/metabolismo , Fosfatos/metabolismo , Fosforilación/efectos de los fármacos , Fosfatos de Poliisoprenilo/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/metabolismo , Pirofosfatasas/genética , Pirofosfatasas/metabolismo
8.
Chin J Traumatol ; 10(3): 180-3, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17535644

RESUMEN

OBJECTIVE: To investigate the mechanism of polymyxin B (PMB) antagonizing the biological activity of lipopolysaccharide (LPS). METHODS: The affinity of PMB for LPS and lipid A was assayed by biosensor, and the neutralization of PMB for LPS (2 ng/ml) was detected by kinetic turbidimetric limulus test. The releases of TNF-alpha and IL-6 in murine peritoneal macrophages a (PMphi) after exposure to LPS (100 ng/ml) were detected, and the expression levels of TLR4, TNF-alpha and IL-6 mRNA in PMphi induced by LPS (100 ng/ml) were measured by RT-PCR. RESULTS: PMB had high-affinity to LPS and lipid A with dissociation equilibrium constants of 18.9 nmol/L and 11.1 nmol/L, respectively, and neutralized LPS in a dose-dependent manner. Furthermore, PMB could markedly inhibit the expressions of TLR4, TNF-alpha and IL-6 mRNA and the release of cycokines in LPS-stimulated murine peritoneal macrophages. CONCLUSIONS: PMB neutralizes LPS and inhibites the expression and release of cycokines in macrophages, in which the affinity of PMB for lipid A plays an important role.


Asunto(s)
Lipopolisacáridos/antagonistas & inhibidores , Polimixina B/farmacología , Animales , Citocinas/análisis , Prueba de Limulus , Lípido A/antagonistas & inhibidores , Macrófagos/química , Ratones
9.
Anesth Analg ; 104(2): 378-83, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17242095

RESUMEN

BACKGROUND: Lipid A, the toxic moiety of endotoxin, is linked to multiple complications after cardiac surgery, including fever, vasodilation, and pulmonary and renal dysfunction. The lipid A antagonist eritoran (or E5564) prevents endotoxin-induced systemic inflammation in animals and humans. In this study we assessed the safety of eritoran administration in patients undergoing cardiac surgery and obtained preliminary efficacy data for the prophylaxis of endotoxin-mediated surgical complications. METHODS: A double-blind, randomized, ascending-dose, placebo-controlled study was conducted at nine hospitals. Patients undergoing coronary artery bypass graft and/or cardiac valvular surgery with cardiopulmonary bypass were enrolled. Patients received a 4-h infusion of placebo (n = 78) vs 2 mg (n = 24), 12 mg (n = 26), or 28 mg (n = 24) of eritoran initiated approximately 1 h before cardiopulmonary bypass. RESULTS: No significant safety concerns were identified with continuous safety monitoring, and enrollment continued to the highest prespecified dose (28 mg). No statistically significant differences were observed in most variables related to systemic inflammation or organ dysfunction/injury. CONCLUSIONS: This Phase II safety study suggests that the administration of the novel lipid A antagonist, eritoran, is not associated with overt toxicity in cardiac surgical patients. Blocking lipid A with eritoran does not appear to confer any clear benefit to elective cardiac surgical patients.


Asunto(s)
Puente Cardiopulmonar , Lípido A/análogos & derivados , Lípido A/antagonistas & inhibidores , Anciano , Procedimientos Quirúrgicos Cardíacos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Femenino , Humanos , Lípido A/administración & dosificación , Lípido A/sangre , Masculino , Persona de Mediana Edad , Cuidados Posoperatorios , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/prevención & control , Estudios Prospectivos
10.
Carbohydr Res ; 341(7): 811-22, 2006 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-16530740

RESUMEN

Glucose analogues 6, 12, 17b, 19a, and 19b of E5564 were synthesized, and their LPS-antagonistic activities were measured. The antagonistic activities (IC(50)) on LPS-induced TNFalpha production of these five compounds toward human whole blood were 72.8, 3.0, 0.9, 7.5, and 1.4nM, respectively. Inhibitory doses (ID(50)) of compounds 12, 17b, 19a, and 19b on TNFalpha production induced by co-injection of galactosamine and LPS in C3H/HeN mice in vivo were measured. The values of these compounds were 0.9, ND (not determined), 1.6, and 0.9mg/kg, respectively.


Asunto(s)
Glucosa/análogos & derivados , Glucosa/biosíntesis , Lípido A/análogos & derivados , Lípido A/antagonistas & inhibidores , Animales , Sangre/efectos de los fármacos , Sangre/metabolismo , Ensayo de Inmunoadsorción Enzimática , Galactosamina/administración & dosificación , Galactosamina/farmacología , Humanos , Concentración 50 Inhibidora , Inyecciones Intravenosas , Lípido A/administración & dosificación , Lípido A/farmacología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Estructura Molecular , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Cell Mol Immunol ; 3(1): 21-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16549045

RESUMEN

Septic shock is caused by Gram-negative bacterial infection. Lipopolysaccharide (LPS) is the bioactive molecule present on the outer membrane of the Gram-negative bacteria. It is generally thought that LPS interacts with sensors on the host cell membrane to activate the intracellular signaling pathway resulting in the overproduction of cytokines such as TNF-alpha. This causes inflammation and ultimately, septic shock. Lipid A is the pharmacophore of the LPS molecule. Thus, developing bio-molecules which are capable of binding LPS at high affinity, especially to the lipid A moiety is an efficient way to neutralize the LPS toxicity. Factor C, a serine protease in the horseshoe crab ameobocytes, is sensitive to trace levels of LPS. We have derived Sushi peptides from the LPS-binding domains of Factor C. Our earlier study showed that the Sushi peptides inhibit LPS-induced septic shock in mice. Here, we demonstrate that the molecular interaction between LPS and Sushi 1 peptide is supported by the hydrophobic interaction between the lipid tail of LPS and Sushi 1 peptide. Furthermore, in the presence of LPS, the peptide transitions from a random structure into an alpha-helical conformation and it disrupts LPS aggregates, hence, neutralizing the LPS toxicity.


Asunto(s)
Precursores Enzimáticos/química , Lípido A/química , Péptidos/química , Pseudomonas/metabolismo , Serina Endopeptidasas/química , Animales , Proteínas de Artrópodos , Sitios de Unión , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/uso terapéutico , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/metabolismo , Humanos , Lípido A/antagonistas & inhibidores , Lípido A/metabolismo , Péptidos/metabolismo , Péptidos/uso terapéutico , Unión Proteica , Estructura Secundaria de Proteína , Pseudomonas/patogenicidad , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/uso terapéutico , Choque Séptico/tratamiento farmacológico , Choque Séptico/metabolismo , Choque Séptico/microbiología , Relación Estructura-Actividad
12.
J Antibiot (Tokyo) ; 59(1): 35-43, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16568717

RESUMEN

Compounds that inactivate lipopolysaccharide (LPS) activity have the potential of being new anti-inflammatory agents. Therefore, we searched among microbial secondary metabolites for compounds that inhibited LPS-stimulated adhesion between human umbilical vein endothelial cells (HUVEC) and HL-60 cells. By this screening, we found a cyclic lipopeptide surfactin from the culture broth of Bacillus sp. BML752-121F2 to be inhibitory. The addition of the surfactin prior to the LPS stimulation decreased HL-60 cell-HUVEC adhesion without showing any cytotoxicity. We confirmed that surfactin inhibited LPS-induced expression of ICAM-1 and VCAM-1 in HUVEC. It also inhibited the cellular adhesion induced by lipid A, the active component of LPS; but it did not inhibit TNF-alpha or IL-1 beta-induced cell adhesion. Then, surfactin was shown to suppress the interaction of lipid A with LPS-binding protein (LBP) that mediates the transport of LPS to its receptors. Finally, surface plasmon resonance (SPR) analysis revealed the surfactin to interact reversibly with lipid A. Thus, this Bacillus surfactin was shown to be an inhibitor of LPS-induced signal transduction, directly interacting with LPS.


Asunto(s)
Lipopolisacáridos/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Bacillus/química , Bacillus/metabolismo , Western Blotting , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colorantes , Medios de Cultivo/química , Células Endoteliales/efectos de los fármacos , Células HL-60/efectos de los fármacos , Humanos , Interleucina-1/farmacología , Lípido A/antagonistas & inhibidores , Lipopéptidos , Resonancia por Plasmón de Superficie , Tensoactivos/farmacología , Azul de Tripano , Factor de Necrosis Tumoral alfa/farmacología
13.
Biomaterials ; 27(9): 1771-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16243394

RESUMEN

Recently [Jessel N, Schwinte P, Donohue R, Lavalle P, Boulmedais F, Darcy R, et al. Pyridylamino-beta-cyclodextrin as a molecular chaperone for lipopolysaccharide embedded in a multilayered polyelectrolyte architecture. Adv Funct Mater 2004;14:963-9], we demonstrated the biological activity of a lipopolysaccharide from Escherichia coli incorporated into layer-by-layer films made of poly (l-lysine) and poly (l-glutamic acid) and containing a polycationic beta-cyclodextrin (CD) with chaperone properties. Here we develop innovative architectures containing a complex made of a charged beta-cyclodextrin and a lipid A antagonist (LAA) as potential systems for local endotoxin antagonistic activity. We examine the biological activity of these architectures. The CD-LAA complex adsorbed on top, or embedded into the polyelectrolyte films keeps its LPS antagonistic activity on both murine and human macrophages for at least 24h.


Asunto(s)
Lípido A/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Poliaminas/farmacología , Animales , Células Cultivadas , Humanos , Lípido A/química , Ratones , Ratones Endogámicos C3H , Péptidos/química , Poliaminas/química , Polielectrolitos , Espectrometría Raman , beta-Ciclodextrinas/química
14.
Immunol Lett ; 100(2): 120-9, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16154492

RESUMEN

We have reported earlier that purified preparations of sheep fetal hemoglobin, but not adult hemoglobin, in concert with non-stimulatory doses of lipopolysaccharide (LPS) (lipid A), act cooperatively to regulate in vitro production of a number of cytokines, including TNFalpha, TGFbeta and IL-6 from murine and human leukocytes. Following in vivo treatment of mice with the same combination of hemoglobin and LPS, harvested spleen or peritoneal cells showed a similar augmented capacity to release these cytokines into culture supernatants. We report below that genetically cloned gamma-chain of human or sheep fetal hemoglobin, but not cloned alpha- or beta-chains, can produce this cooperative effect, as indeed can HPLC purified, heme-free, gamma-chains derived from cord blood fetal hemoglobin, and that purified haptoglobin completely abolishes the cooperative interaction.


Asunto(s)
Hemoglobina Fetal/inmunología , Globinas/inmunología , Lipopolisacáridos/inmunología , Linfocitos/efectos de los fármacos , Bazo/efectos de los fármacos , Factores de Edad , Secuencia de Aminoácidos , Animales , Clonación Molecular , Cricetinae , Relación Dosis-Respuesta a Droga , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Globinas/biosíntesis , Globinas/química , Haptoglobinas/farmacología , Humanos , Interleucina-6/biosíntesis , Lípido A/administración & dosificación , Lípido A/antagonistas & inhibidores , Lípido A/inmunología , Lipopolisacáridos/administración & dosificación , Ratones , Datos de Secuencia Molecular , Ovinos , Bazo/citología , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis
15.
J Med Chem ; 48(7): 2589-99, 2005 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15801849

RESUMEN

Lipopolysaccharides (LPS), otherwise termed "endotoxins", are outer membrane constituents of Gram-negative bacteria. Lipopolysaccharides play a key role in the pathogenesis of "septic shock", a major cause of mortality in the critically ill patient. Therapeutic options aimed at limiting downstream systemic inflammatory processes by targeting lipopolysaccharide do not exist at the present time. We have defined the pharmacophore necessary for small molecules to specifically bind and neutralize LPS and, using animal models of sepsis, have shown that the sequestration of circulatory LPS by small molecules is a therapeutically viable strategy. In this paper, the interactions of a series of acylated homologated spermine compounds with LPS have been characterized. The optimal acyl chain length for effective sequestration of LPS was identified to be C(16) for the monoacyl compounds. The most promising of these compounds, 4e, binds LPS with an ED(50) of 1.37 muM. Nitric oxide production in murine J774A.1 cells, as well as TNF-alpha in human blood, is inhibited in a dose-dependent manner by 4e at concentrations orders of magnitude lower than toxic doses. Administration of 4e to d-galactosamine-sensitized mice challenged with supralethal doses of LPS provided significant protection against lethality. Potent antiendotoxic activity, low toxicity, and ease of synthesis render this class of compounds candidate endotoxin-sequestering agents of potential significant therapeutic value.


Asunto(s)
Amidas/síntesis química , Lipopolisacáridos/antagonistas & inhibidores , Espermina/análogos & derivados , Espermina/síntesis química , Amidas/química , Amidas/farmacología , Amidas/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Escherichia coli , Femenino , Hemólisis , Humanos , Técnicas In Vitro , Lípido A/antagonistas & inhibidores , Lípido A/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Sepsis/mortalidad , Sepsis/prevención & control , Espermina/química , Espermina/farmacología , Espermina/toxicidad , Relación Estructura-Actividad , Propiedades de Superficie , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
FEMS Immunol Med Microbiol ; 43(1): 91-8, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15607641

RESUMEN

The protein-bound polysaccharide isolated from basidiomycetes (PSK) is a biological response modifier capable of exhibiting various biological activities, such as antitumor and antimicrobial effects. In the present study, we found that PSK suppressed interleukin (IL)-6 production in murine peritoneal macrophages stimulated with endotoxic lipopolysaccharide (LPS) and its synthetic lipid A (compound 506). Nitric oxide production and p38 mitogen-associated protein kinase phosphorylation induced in a murine macrophage cell line, J774-A1, by LPS and compound 506 were also inhibited by PSK. Further, PSK distinctly suppressed nuclear factor-kappaB activation in Ba/F3 cells expressing mouse Toll-like receptor 4 and MD-2, following stimulation with LPS and compound 506, however, not with Taxol. These PSK-induced inhibitory activities were caused by inhibition of the physical associations of LPS with LPS-binding protein (LBP) and CD14. PSK also protected mice from LPS-induced lethality, presumably by down-regulating IL-6 and tumor necrosis factor-alpha concentrations in serum. These findings indicate that PSK, which also has an ability to regulate LBP/CD14 functions, may be useful for clinical control of endotoxic sepsis.


Asunto(s)
Proteínas de Fase Aguda/antagonistas & inhibidores , Antitoxinas/farmacología , Basidiomycota/química , Proteínas Portadoras/antagonistas & inhibidores , Lípido A/antagonistas & inhibidores , Receptores de Lipopolisacáridos/fisiología , Glicoproteínas de Membrana/antagonistas & inhibidores , Proteoglicanos/farmacología , Receptores Inmunológicos/antagonistas & inhibidores , Proteínas de Fase Aguda/inmunología , Animales , Proteínas Portadoras/inmunología , Células Cultivadas , Interleucina-6/biosíntesis , Lípido A/síntesis química , Activación de Macrófagos , Macrófagos Peritoneales , Masculino , Glicoproteínas de Membrana/inmunología , Ratones , FN-kappa B/análisis , Óxido Nítrico/biosíntesis , Proteoglicanos/aislamiento & purificación , Proteínas Quinasas p38 Activadas por Mitógenos/análisis
17.
Int Immunopharmacol ; 4(4): 527-37, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15099530

RESUMEN

Lipopolysaccharide (LPS [endotoxin]), a structural component of gram-negative bacteria, is implicated in the pathogenesis of septic shock. Lipid A is an evolutionarily conserved region of LPS that has been identified as the toxic component of LPS. Therapeutic strategies for the treatment of septic shock in humans are currently focused on neutralization of LPS. Here, the anti-endotoxin activity of BNEP, a synthetic peptide derived from the human bactericidal/permeability-increasing protein (BPI; aa 148-161) was investigated in vitro and in experimental animal endotoxemia models in vivo. The ability of BNEP to bind LPS from Escherichia coli O55:B5 and lipid A from Salmonella Re 595 was tested using an affinity sensor assay, and its ability to neutralize LPS was tested using a sensitive Limulus amebocyte lysate (LAL) assay. Polymyxin B (PMB) was used as the positive control in the in vitro experiments and in mouse experiments. We found that BNEP and PMB bound LPS with a similar affinity (Kd values of 25.4 and 25.8 nM, respectively). In contrast, BNEP bound lipid A with a slightly lower affinity than that of PMB (Kd values of 8 and 5.6 nM, respectively). The exact capacity of BNEP binding to LPS was approximately 0.53 microg peptide per 1 ng of LPS, as shown by affinity sensor assay. The LAL test showed that 256 microg of BNEP almost completely neutralized 2 ng LPS. In vivo, mice were randomized, intravenously injected with BNEP (0.5-10 mg/kg) or 1 mg/kg PMB, and then lethally challenged with 20 mg/kg LPS. We found that 5 mg/kg BNEP significantly protected mice from LPS challenge. In an endotoxemia rat model, animals were co-treated with 5 or 10 mg/kg BNEP and 10 mg/kg LPS via cardiac catheter. BNEP treatment resulted in significant reduction of tumor necrosis factor alpha (TNF-alpha) and IL-6, compared with LPS-only control animals. In addition, 10 mg/kg BNEP-treated animals showed a significant decrease in plasma endotoxin levels in comparison to animals treated with LPS alone. These results provide evidence that BNEP effectively neutralizes LPS in vitro and in vivo, and could protect animals from the lethal effects of LPS via decreasing plasma endotoxin and proinflammatory cytokines. Our work suggests that this peptide is worthy of further investigation as a possible novel treatment for septic shock.


Asunto(s)
Proteínas Sanguíneas/química , Lipopolisacáridos/antagonistas & inhibidores , Proteínas de la Membrana/química , Péptidos/farmacología , Animales , Péptidos Catiónicos Antimicrobianos , Relación Dosis-Respuesta a Droga , Endotoxemia/tratamiento farmacológico , Femenino , Interleucina-6/antagonistas & inhibidores , Lípido A/antagonistas & inhibidores , Lípido A/química , Lípido A/metabolismo , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Unión Proteica , Ratas , Ratas Wistar , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
18.
J Clin Pharmacol ; 43(7): 735-42, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12856387

RESUMEN

E5564, a structural analog of the lipid A portion of lipopolysaccharide (LPS), is a potent antagonist of the biochemical and physiologic effects of LPS in several in vitro and in vivo models and is currently under clinical development as a possible therapeutic for the treatment of sepsis and septic shock. The objectives of this study were to (1) assess the safety and tolerability of E5564 following a 30-minute intravenous (i.v.) infusion, (2) evaluate the pharmacokinetic profile of E5564, and (3) measure the ability of E5564 to block LPS stimulation ex vivo in blood taken from subjects up to 8 hours after ending the infusion. Healthy male volunteers (n = 7/dose group) were randomly assigned to each of four dose levels (350, 1000, 2000, or 3500 micrograms). Within each dose group, 5 subjects received drug and 2 received placebo. E5564 or matching placebo was administered by a 30-minute infusion, and blood samples were collected at predetermined time points. All doses of E5564 were demonstrated to be safe and well tolerated. E5564 plasma concentrations were determined using a validated LC/MS/MS method. The Cmax and AUC of E5564 increased in a dose-proportional manner. E5564 pharma-cokinetics were characterized by a slow clearance (0.67-0.95 mL/h/kg), a small volume of distribution (41-54 mL/kg), and a relatively long elimination half-life (42-51 h). As measured in the ex vivo assay, E5564 inhibited LPS-induced tumor necrosis factor-alpha (TNF-alpha) in a dose-dependent manner, and at the higher doses (2 and 3.5 mg), antagonistic activity was measurable up to 8 hours postinfusion. E5564 lacked LPS-like agonist activity at doses up to 3.5 mg. Taken together, we believe that E5564 is a safe, potent antagonist of LPS in blood and will likely benefit patients in the treatment of LPS-related diseases.


Asunto(s)
Lípido A , Lípido A/análogos & derivados , Lípido A/antagonistas & inhibidores , Adulto , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Semivida , Humanos , Inyecciones Intravenosas , Lípido A/administración & dosificación , Lípido A/efectos adversos , Lípido A/farmacocinética , Lípido A/farmacología , Masculino , Tasa de Depuración Metabólica , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Med Microbiol ; 49(2): 127-138, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10670563

RESUMEN

The saccharide constituents of lipopolysaccharides (LPS) of Proteus spp. vary with the strain and contain unique components about which little is known. The biological activities of LPS and lipid A from S- and R-forms of 10 Proteus strains were examined. LPS from all S-form Proteus strains was lethal to D-(+)-galactosamine (GalN)-loaded, LPS-responsive, C3H/HeN mice, but not to LPS-hypo-responsive C3H/HeJ mice. P. vulgaris 025 LPS evoked strong anaphylactoid reactions in N-acetylmuramyl-L-alanyl-D-isoglutamine (MDP)-primed C3H/HeJ mice. LPS from S- and R-form Proteus strains induced production of nitric oxide (NO) and tumour necrosis factor (TNF) by macrophages isolated from C3H/HeN but not C3H/HeJ mice. Lipid A from Proteus strains also induced NO and TNF production, although lipid A was less potent than LPS. The effects of LPS were mainly dependent on CD14; LPS-induced NO and TNF production in CD14+ J774.1 cells was significantly greater than in CD14-J7.DEF.3 cells. All LPS from Proteus strains, and especially from P. vulgaris 025, exhibited higher anti-complementary activity than LPS from Escherichia coli or Pseudomonas aeruginosa. Polymyxin B inactivated proteus LPS in a dose-dependent manner, but these LPS preparations were more resistant to polymyxin B than E. coli LPS. CAP18(109-135), a granulocyte-derived peptide, inhibited proteus LPS endotoxicity only when the LPS:CAP18(109-135) ratio was appropriate, which suggests that CAP18(109-135) acts through a different mechanism than polymyxin B. The results indicate that LPS from Proteus spp. are potently endotoxic, but that the toxicity is different from that of LPS from E. coli or Salmonella spp. and even varies among different Proteus strains. The variation in biological activities among proteus LPS may be due to unique components within the respective LPS.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Proteínas Portadoras/farmacología , Lipopolisacáridos/toxicidad , Fragmentos de Péptidos/farmacología , Polimixina B/farmacología , Proteus/efectos de los fármacos , Proteus/patogenicidad , Acetilmuramil-Alanil-Isoglutamina/administración & dosificación , Secuencia de Aminoácidos , Anafilaxia/inducido químicamente , Animales , Secuencia de Carbohidratos , Proteínas Portadoras/química , Catelicidinas , Proteínas Inactivadoras de Complemento/farmacología , Femenino , Galactosamina/administración & dosificación , Lípido A/antagonistas & inhibidores , Lípido A/toxicidad , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/antagonistas & inhibidores , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Óxido Nítrico/biosíntesis , Proteus/metabolismo , Proteus mirabilis/efectos de los fármacos , Proteus mirabilis/metabolismo , Proteus mirabilis/patogenicidad , Proteus vulgaris/efectos de los fármacos , Proteus vulgaris/metabolismo , Proteus vulgaris/patogenicidad , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Br J Pharmacol ; 127(4): 853-62, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10433491

RESUMEN

1. The major pathological responses to Gram-negative bacterial sepsis are triggered by endotoxin or lipopolysaccharide. As endotoxin is shed from the bacterial outer membrane, it induces immunological responses that lead to release of a variety of cytokines and other cellular mediators. As part of a program aimed at developing a therapeutic agent for septic shock, we have developed E5531, a novel synthetic lipopolysaccharide antagonist. 2. As measured by release by tumour necrosis factor-alpha, human monocytes or whole blood can be activated by lipopolysaccharide, lipid A, and lipoteichoic acid (from Gram-positive bacteria). E5531 potently antagonizes activation by all these agents while itself being devoid of agonistic activity. 3. The inhibitory activity of E5531 was dependent on time of addition. When 10 nM E5531 was added simultaneously with lipopolysaccharide or 1 - 3 h before addition of lipopolysaccharide, production of tumour necrosis factor-alpha was inhibited by more than 98%. The addition of E5531 1 h after lipopolysaccharide reduced the efficacy of E5531 by 47%. 4. Antagonistic activity of E5531 was specific for lipopolysaccharide as it was ineffective at inhibiting interferon-gamma mediated NO release of RAW 264.7 cells, phorbor 12-myristate 13-acetate stimulated superoxide anion production in human neutrophils, concanavalin A stimulated mitogenic activity in murine thymocytes and tumor necrosis factor-alpha induced E-selectin expression in human umbilical vein endothelial cells. 5. E5531 as well as MY4, an anti-CD14 antibody, inhibited radiolabelled lipopolysaccharide binding in human monocytes. 6. These results support our contention that E5531 is a potent antagonist of lipopolysaccharide-induced release of tumour necrosis factor-alpha and other cellular mediators and may be an effective therapeutic agent for human septic shock due to Gram-negative bacteria.


Asunto(s)
Lípido A/análogos & derivados , Lipopolisacáridos/antagonistas & inhibidores , Selectina E/biosíntesis , Humanos , Interferón gamma/farmacología , Lípido A/antagonistas & inhibidores , Lípido A/farmacología , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Óxido Nítrico/biosíntesis , Superóxidos/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA