Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 83(9): 3555-67, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26099588

RESUMEN

We previously reported that Neisseria meningitidis internalization into human brain microvasocular endothelial cells (HBMEC) was triggered by the influx of extracellular L-glutamate via the GltT-GltM L-glutamate ABC transporter, but the underlying mechanism remained unclear. We found that the ΔgltT ΔgltM invasion defect in assay medium (AM) was alleviated in AM without 10% fetal bovine serum (FBS) [AM(-S)]. The alleviation disappeared again in AM(-S) supplemented with 500 µM glutamate. Glutamate uptake by the ΔgltT ΔgltM mutant was less efficient than that by the wild-type strain, but only upon HBMEC infection. We also observed that both GltT-GltM-dependent invasion and accumulation of ezrin, a key membrane-cytoskeleton linker, were more pronounced when N. meningitidis formed larger colonies on HBMEC under physiological glutamate conditions. These results suggested that GltT-GltM-dependent meningococcal internalization into HBMEC might be induced by the reduced environmental glutamate concentration upon infection. Furthermore, we found that the amount of glutathione within the ΔgltT ΔgltM mutant was much lower than that within the wild-type N. meningitidis strain only upon HBMEC infection and was correlated with intracellular survival. Considering that the L-glutamate obtained via GltT-GltM is utilized as a nutrient in host cells, l-glutamate uptake via GltT-GltM plays multiple roles in N. meningitidis internalization into HBMEC.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Células Endoteliales/microbiología , Ácido Glutámico/metabolismo , Neisseria meningitidis/patogenicidad , Infecciones por Neisseriaceae/metabolismo , Western Blotting , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Neisseria meningitidis/metabolismo
2.
PLoS Pathog ; 8(4): e1002597, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496641

RESUMEN

LPS-activated neutrophils secrete IL-1ß by activation of TLR-4. Based on studies in macrophages, it is likely that ROS and lysosomal destabilization regulated by Syk activation may also be involved. Since neutrophils have abundant expression of the ITIM-containing co-receptor CEACAM1 and Gram-negative bacteria such as Neisseria utilize CEACAM1 as a receptor that inhibits inflammation, we hypothesized that the overall production of IL-1ß in LPS treated neutrophils may be negatively regulated by CEACAM1. We found that LPS treated neutrophils induced phosphorylation of Syk resulting in the formation of a complex including TLR4, p-Syk, and p-CEACAM1, which in turn, recruited the inhibitory phosphatase SHP-1. LPS treatment leads to ROS production, lysosomal damage, caspase-1 activation and IL-1ß secretion in neutrophils. The absence of this regulation in Ceacam1⁻/⁻ neutrophils led to hyper production of IL-1ß in response to LPS. The hyper production of IL-1ß was abrogated by in vivo reconstitution of wild type but not ITIM-mutated CEACAM1 bone marrow stem cells. Blocking Syk activation by kinase inhibitors or RNAi reduced Syk phosphorylation, lysosomal destabilization, ROS production, and caspase-1 activation in Ceacam1⁻/⁻ neutrophils. We conclude that LPS treatment of neutrophils triggers formation of a complex of TLR4 with pSyk and pCEACAM1, which upon recruitment of SHP-1 to the ITIMs of pCEACAM1, inhibits IL-1ß production by the inflammasome. Thus, CEACAM1 fine-tunes IL-1ß production in LPS treated neutrophils, explaining why the additional utilization of CEACAM1 as a pathogen receptor would further inhibit inflammation.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Interleucina-1beta/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/farmacología , Complejos Multiproteicos/metabolismo , Activación Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Lisosomas/genética , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Neisseria/inmunología , Neisseria/metabolismo , Infecciones por Neisseriaceae/genética , Infecciones por Neisseriaceae/inmunología , Infecciones por Neisseriaceae/metabolismo , Infecciones por Neisseriaceae/patología , Activación Neutrófila/genética , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo
3.
PLoS Pathog ; 5(10): e1000629, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19851451

RESUMEN

The bacterial PorB porin, an ATP-binding beta-barrel protein of pathogenic Neisseria gonorrhoeae, triggers host cell apoptosis by an unknown mechanism. PorB is targeted to and imported by host cell mitochondria, causing the breakdown of the mitochondrial membrane potential (DeltaPsi(m)). Here, we show that PorB induces the condensation of the mitochondrial matrix and the loss of cristae structures, sensitizing cells to the induction of apoptosis via signaling pathways activated by BH3-only proteins. PorB is imported into mitochondria through the general translocase TOM but, unexpectedly, is not recognized by the SAM sorting machinery, usually required for the assembly of beta-barrel proteins in the mitochondrial outer membrane. PorB integrates into the mitochondrial inner membrane, leading to the breakdown of DeltaPsi(m). The PorB channel is regulated by nucleotides and an isogenic PorB mutant defective in ATP-binding failed to induce DeltaPsi(m) loss and apoptosis, demonstrating that dissipation of DeltaPsi(m) is a requirement for cell death caused by neisserial infection.


Asunto(s)
Apoptosis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Porinas/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Proteínas Bacterianas/fisiología , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Modelos Biológicos , Neisseria gonorrhoeae/química , Neisseria gonorrhoeae/patogenicidad , Neisseria gonorrhoeae/fisiología , Infecciones por Neisseriaceae/metabolismo , Infecciones por Neisseriaceae/patología , Porinas/metabolismo , Porinas/fisiología
4.
Glia ; 53(6): 583-92, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16419089

RESUMEN

There is growing appreciation that resident glial cells can initiate and/or regulate inflammation following trauma or infection in the central nervous system (CNS). We have previously demonstrated the ability of microglia and astrocytes, resident glial cells of the CNS, to respond to bacterial pathogens by rapid production of inflammatory mediators. However, inflammation within the brain parenchyma is notably absent during some chronic bacterial infections in humans and nonhuman primates. In the present study, we demonstrate the ability of the immunosuppressive cytokine, interleukin-10 (IL-10), to inhibit inflammatory immune responses of primary microglia and astrocytes to B. burgdorferi and N. meningitidis, two disparate gram negative bacterial species that can cross the blood-brain barrier in humans. Importantly, we demonstrate that these organisms induce the delayed production of significant quantities of IL-10 by both microglia and astrocytes. Furthermore, we demonstrate that such production occurs independent of the actions of bacterial lipopolysaccharide and is secondary to the autocrine or paracrine actions of other glia-derived soluble mediators. The late onset of IL-10 production by resident glia following activation, the previously documented expression of specific receptors for this cytokine on microglia and astrocytes, and the ability of IL-10 to inhibit bacterially induced immune responses by these cells, suggest a mechanism by which resident glial cells can limit potentially damaging inflammation within the CNS in response to invading pathogens, and could explain the suppression of inflammation seen within the brain parenchyma during chronic bacterial infections.


Asunto(s)
Borrelia burgdorferi/inmunología , Encefalitis/inmunología , Tolerancia Inmunológica/inmunología , Interleucina-10/inmunología , Neisseria meningitidis/inmunología , Neuroglía/inmunología , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Infecciones por Borrelia/inmunología , Infecciones por Borrelia/metabolismo , Infecciones por Borrelia/fisiopatología , Línea Celular Transformada , Células Cultivadas , Quimiotaxis/inmunología , Encefalitis/metabolismo , Encefalitis/microbiología , Encefalitis/fisiopatología , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/fisiopatología , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C3H , Microglía/inmunología , Infecciones por Neisseriaceae/inmunología , Infecciones por Neisseriaceae/metabolismo , Infecciones por Neisseriaceae/fisiopatología , Neuroglía/microbiología , Comunicación Paracrina/inmunología , Factores de Tiempo
5.
Proteomics ; 5(8): 2048-55, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15852346

RESUMEN

Neisseria meningitidis is the most common cause of meningitis and causes epidemic outbreaks. One trait of N. meningitidis, which is associated with most of the currently recognized virulence determinants, is the presence of phase-variable genes that are suspected to enhance its ability to cause an invasive disease. To detect the immune responses to phase-variable expressed proteins, we applied protein microarray technology for the screening of meningitis patient sera. We amplified all 102 known phase-variable genes from N. meningitidis serogroup B strain MC58 by polymerase chain reaction and subcloned them for expression in Escherichia coli. With this approach, we were able to express and purify 67 recombinant proteins representing 66% of the annotated genes. These were spotted robotically onto coated glass slides to generate protein microarrays, which were screened using 20 sera of patients suffering from meningitis, as well as healthy controls. From these screening experiments, 47 proteins emerged as immunogenic, exhibiting a variable degree of seroreactivity with some of the patient sera. Nine proteins elicited an immune response in more than three patients, with one of them, the phase-variable opacity protein OpaV (NMB0442), showing responses in 11 patient sera. This is the first time that protein microarray technology has been applied for the investigation of genetic phase variation in pathogens. The identification of disease-specific proteins is a significant target in biomedical research, as such proteins may have medical, diagnostic, and commercial potential as disease markers.


Asunto(s)
Proteínas Bacterianas/análisis , Neisseria meningitidis/genética , Infecciones por Neisseriaceae/diagnóstico , Análisis por Matrices de Proteínas/métodos , Biomarcadores/sangre , Western Blotting , Escherichia coli/genética , Histidina/química , Procesamiento de Imagen Asistido por Computador , Neisseria meningitidis/clasificación , Neisseria meningitidis/aislamiento & purificación , Neisseria meningitidis/metabolismo , Infecciones por Neisseriaceae/sangre , Infecciones por Neisseriaceae/metabolismo , Técnicas de Amplificación de Ácido Nucleico , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Serotipificación
6.
J Cell Biol ; 156(6): 951-7, 2002 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-11901164

RESUMEN

The Neisseria type IV pilus promotes bacterial adhesion to host cells. The pilus binds CD46, a complement-regulatory glycoprotein present on nucleated human cells (Källström et al., 1997). CD46 mutants with truncated cytoplasmic tails fail to support bacterial adhesion (Källström et al., 2001), suggesting that this region of the molecule also plays an important role in infection. Here, we report that infection of human epithelial cells by piliated Neisseria gonorrhoeae (GC) leads to rapid tyrosine phosphorylation of CD46. Studies with wild-type and mutant tail fusion constructs demonstrate that Src kinase phosphorylates tyrosine 354 in the Cyt2 isoform of the CD46 cytoplasmic tail. Consistent with these findings, infection studies show that PP2, a specific Src family kinase inhibitor, but not PP3, an inactive variant of this drug, reduces the ability of epithelial cells to support bacterial adhesion. Several lines of evidence point to the role of c-Yes, a member of the Src family of nonreceptor tyrosine kinases, in CD46 phosphorylation. GC infection causes c-Yes to aggregate in the host cell cortex beneath adherent bacteria, increases binding of c-Yes to CD46, and stimulates c-Yes kinase activity. Finally, c-Yes immunoprecipitated from epithelial cells is able to phosphorylate the wild-type Cyt2 tail but not the mutant derivative in which tyrosine 354 has been substituted with alanine. We conclude that GC infection leads to rapid tyrosine phosphorylation of the CD46 Cyt2 tail and that the Src kinase c-Yes is involved in this reaction. Together, the findings reported here and elsewhere strongly suggest that pilus binding to CD46 is not a simple static process. Rather, they support a model in which pilus interaction with CD46 promotes signaling cascades important for Neisseria infectivity.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Celular/genética , Células Epiteliales/metabolismo , Fimbrias Bacterianas/metabolismo , Glicoproteínas de Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tirosina/metabolismo , Antígenos CD/genética , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/microbiología , Fimbrias Bacterianas/ultraestructura , Técnica del Anticuerpo Fluorescente , Humanos , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/genética , Neisseria gonorrhoeae/citología , Infecciones por Neisseriaceae/genética , Infecciones por Neisseriaceae/metabolismo , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-yes , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
7.
Int J Antimicrob Agents ; 17(6): 483-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11397619

RESUMEN

Over the last decade or so there has been a growing interest in routes of antimicrobial administration other than by the conventional intravenous route for institutionalized patients and for some outpatients. Both oral (PO) and intramuscular (IM) routes of administration are less costly than giving antimicrobial agents by vein (IV). In addition, fewer complications such as catheter-related sepsis and phlebitis are associated with non-IV routes of administration. Furthermore, a reduced-dosage, reduced-volume IM administration of ceftriaxone may provide a tolerable route of administration and equivalent bactericidal activities compared with higher dose IV ceftriaxone. The purpose of this study was to determine the time that the drug concentration remained in excess of the minimum inhibitory concentration (MIC) (T > MIC) and the duration of bactericidal activities of ceftriaxone one gram administered IV, ceftriaxone 250 mg given IM and cefixime 400 mg given orally against clinical isolates of Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis in adult volunteers. Single doses of each agent were administered and serum concentrations were collected over the standard dosing period of 24 h for all study regimens. Ceftriaxone, regardless of route of administration and dose, resulted in bactericidal activities and T > MIC for 100% of the dosing period for S. pneumoniae, H. influenzae, and M. catarrhalis. Cefixime maintained at least 50% T > MIC and bactericidal activity against both isolates each of H. influenzae and M. catarrhalis. Against both isolates of S. pneumoniae, cefixime achieved T > MIC for at least 50% of the dosing period, but did not maintain bactericidal activity. Reduced dose ceftriaxone given IM seems to be a viable alternative to ceftriaxone IV if the pathogen, susceptibility and infection site are known. Based on T > MIC exceeding 50% of the dosing interval, cefixime would be considered an effective alternative to IV therapy against common respiratory tract pathogens. Clinical studies need to be conducted to confirm these findings.


Asunto(s)
Cefixima/farmacología , Ceftriaxona/farmacología , Cefalosporinas/farmacología , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Administración Oral , Adulto , Actividad Bactericida de la Sangre , Cefixima/administración & dosificación , Cefixima/farmacocinética , Ceftriaxona/administración & dosificación , Ceftriaxona/farmacocinética , Cefalosporinas/administración & dosificación , Cefalosporinas/farmacocinética , Infecciones Comunitarias Adquiridas/metabolismo , Estudios Cruzados , Femenino , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/aislamiento & purificación , Humanos , Inyecciones Intramusculares , Inyecciones Intravenosas , Masculino , Moraxella catarrhalis/efectos de los fármacos , Moraxella catarrhalis/aislamiento & purificación , Infecciones por Neisseriaceae/tratamiento farmacológico , Infecciones por Neisseriaceae/metabolismo , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/aislamiento & purificación
8.
Jpn J Antibiot ; 54(10): 532-40, 2001 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-11771335

RESUMEN

Clinical studies of ceftriaxone (CTRX) were performed at a dose of 40 mg/kg once daily to evaluate its pharmacokinetics, and clinical and bacteriological efficacies in pediatric patients with respiratory tract infections. The following results were obtained. 1. Of 45 patients, clinical responses to CTRX were excellent in 34 (75.6%), good in 9 (20.0%) and poor in 2 (4.4%), indicating the overall efficacy rate of 95.6%. 2. Haemophilus influenzae (23 strains), Streptococcus pneumoniae (20 strains) and Moraxella catarrhalis (17 strains) were isolated from the patients as the main causative organisms. MIC90 of CTRX against these detected bacteria was < or = 0.06 microgram/ml with H. influenzae [beta-lactamase (-)/ABPC (S)], 0.25 microgram/ml with H. influenzae (BLNAR), 0.05 microgram/ml with PSSP, 1.0 microgram/ml with PISP/PRSP and 2.0 micrograms/ml with M. catarrhalis, respectively. 3. The eradication rate of causative organisms was 90.0% (27/30). 4. Serum levels of CTRX after administration of a 1-hour intravenous drip infusion of 40 mg/kg were investigated in 12 patients. Mean serum level at 24 hours after the administration was 9.4 +/- 2.8 micrograms/ml, which covered the level of MIC90 throughout the 24 hours. 5. No adverse reactions related to CTRX were observed. As the approved dosage of CTRX in pediatric patients is twice daily, while it is once daily in adults, there have been few reports on the efficacy of once-daily CTRX in pediatrics. According to the results of the study, it is suggested that once-daily CTRX for the pediatric patients with respiratory tract infections is useful. Further studies might be required to establish outpatient parenteral antibiotic therapy (OPAT) in pediatric infections.


Asunto(s)
Ceftriaxona/administración & dosificación , Cefalosporinas/administración & dosificación , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Ceftriaxona/farmacocinética , Cefalosporinas/farmacocinética , Niño , Preescolar , Esquema de Medicación , Femenino , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/aislamiento & purificación , Humanos , Lactante , Infusiones Intravenosas , Masculino , Moraxella catarrhalis/aislamiento & purificación , Infecciones por Neisseriaceae/tratamiento farmacológico , Infecciones por Neisseriaceae/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/microbiología
9.
J Chemother ; 12(3): 216-22, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10877516

RESUMEN

Cefaclor advanced formulation (cefaclor AF) is an extended-release form of the oral cephalosporin cefaclor. When cefaclor AF 750 mg twice-daily and cefaclor immediate release 500 mg three-times-a-day are compared there is a skew to the right of the pharmacokinetic profile and higher levels are achieved. Based on this pharmacokinetic finding, we examined the relationship between the bacterial susceptibility to cefaclor (MIC), the achieved cefaclor AF serum and sputum concentrations, and in vivo eradication of the bacteria in 36 patients with acute exacerbations of chronic bronchitis. The mean peak concentrations in serum and sputum 5 h after administration were 8.6 microg/ml (95% CI: 8.1 microg/ml - 9.1 microg/ml) and 1.5 microg/ml (95% CI: 1.4 microg/ml - 1.7 microg/ml), respectively. Cefaclor was always detectable 8 h after administration. At post therapy, treatment was successful in 31 (86.1%) patients. Cefaclor concentrations in serum persisted above the MIC for more than 40% of dosing interval in 31 subjects, and those in sputum in 24 patients. Treatment was successful in all subjects with percent of time above the MIC in serum of >40%, whereas the time that levels in sputum stayed above the MIC was not the pharmacodynamic parameter that correlated best with therapeutic efficacy for cefaclor. Our data demonstrate that when cefaclor AF is dosed twice-daily, the in vivo pharmacodynamic susceptibility breakpoint is 8 microg/ml. The good activity and pharmacokinetics of cefaclor AF provide serum concentrations higher than the MIC of Haemophilus influenzae, Streptococcus pneumoniae, and Moraxella catarrhalis for more than 40% of the validated dosing interval. Therefore, it might be considered for first choice treatment of acute exacerbations of chronic bronchitis.


Asunto(s)
Bronquitis/tratamiento farmacológico , Bronquitis/metabolismo , Cefaclor/farmacología , Cefaclor/farmacocinética , Cefalosporinas/farmacología , Cefalosporinas/farmacocinética , Enfermedad Aguda , Bronquitis/microbiología , Cefaclor/administración & dosificación , Cefalosporinas/administración & dosificación , Enfermedad Crónica , Preparaciones de Acción Retardada , Infecciones por Haemophilus/sangre , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/aislamiento & purificación , Humanos , Pruebas de Sensibilidad Microbiana , Moraxella catarrhalis/efectos de los fármacos , Moraxella catarrhalis/aislamiento & purificación , Infecciones por Neisseriaceae/sangre , Infecciones por Neisseriaceae/tratamiento farmacológico , Infecciones por Neisseriaceae/metabolismo , Infecciones Neumocócicas/sangre , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/metabolismo , Esputo/metabolismo , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/aislamiento & purificación , Resultado del Tratamiento
12.
Drugs ; 42 Suppl 3: 34-40, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1726206

RESUMEN

Cefpodoxime proxetil is the orally absorbed ester of cefpodoxime, a new third generation cephalosporin. In the gastrointestinal tract, cefpodoxime proxetil is hydrolysed to cefpodoxime, which has potent antibacterial activity against the major bacterial pathogens involved in lower respiratory tract infections: Haemophilus influenzae, Moraxella (Branhamella) catarrhalis (including beta-lactamase-producing strains), and Streptococcus pneumoniae (including amoxicillin-resistant strains). Six randomised comparative studies in patients with lower respiratory tract infections, 5 of which were large (enrollment of more than 200 patients) and double-blind, examined the efficacy and safety of cefpodoxime proxetil. Cefpodoxime proxetil (at a dosage equivalent to 200mg of cefpodoxime) administered twice daily for 5 to 10 days was similar in clinical and bacteriological efficacy to the following: amoxicillin 500mg 3 times daily in the treatment of community-acquired pneumonia; intramuscular ceftriaxone Ig once daily in the treatment of pulmonary infections in hospitalised patients; and to amoxicillin/clavulanic acid 500/125mg 3 times daily in the treatment of acute exacerbations of chronic bronchitis (AECB). Additionally, a dosage equivalent to 100mg or 200mg of cefpodoxime twice daily was similar in clinical and bacteriological efficacy to amoxicillin 250mg 3 times daily in the treatment of bronchitis (acute or AECB). The adverse events noted with cefpodoxime proxetil administration were similar to those associated with other beta-lactam antibacterials and most commonly involved the gastrointestinal tract and skin or mucous membranes. Thus, cefpodoxime proxetil is a useful addition to the antibacterials available for the treatment of infections of the lower respiratory tract.


Asunto(s)
Bronquitis/etiología , Ceftizoxima/análogos & derivados , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Neisseriaceae/tratamiento farmacológico , Infecciones Neumocócicas/tratamiento farmacológico , Profármacos/uso terapéutico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Anciano , Bronquitis/tratamiento farmacológico , Ceftizoxima/efectos adversos , Ceftizoxima/farmacocinética , Ceftizoxima/uso terapéutico , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae , Humanos , Moraxella catarrhalis , Infecciones por Neisseriaceae/metabolismo , Infecciones Neumocócicas/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Infecciones del Sistema Respiratorio/metabolismo , Streptococcus pneumoniae , Cefpodoxima Proxetilo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA