Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 67(11): e0066523, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37843305

RESUMEN

Acinetobacter baumannii-calcoaceticus complex (ABC) causes severe infections that are difficult to treat due to pre-existing antibiotic resistance. Sulbactam-durlobactam (SUL-DUR) is a targeted ß-lactam/ß-lactamase inhibitor combination antibiotic designed to treat serious infections caused by Acinetobacter, including multidrug- and carbapenem-resistant strains. In a recent global surveillance study of 5,032 ABC clinical isolates collected from 2016 to 2021, less than 2% of ABC isolates had SUL-DUR MIC values >4 µg/mL. Molecular characterization of these isolates confirmed the primary drivers of resistance are metallo-ß-lactamases or penicillin-binding protein 3 (PBP3) mutations, as previously described. In addition, this study shows that certain common PBP3 variants, such as A515V, are insufficient to confer sulbactam resistance and that the efflux of durlobactam by AdeIJK is likely to play a role in a subset of strains.


Asunto(s)
Acinetobacter baumannii , Sulbactam , Sulbactam/farmacología , Sulbactam/uso terapéutico , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/farmacología , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/uso terapéutico , Monobactamas , Pruebas de Sensibilidad Microbiana
2.
Nature ; 597(7878): 698-702, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34526714

RESUMEN

The development of new antibiotics to treat infections caused by drug-resistant Gram-negative pathogens is of paramount importance as antibiotic resistance continues to increase worldwide1. Here we describe a strategy for the rational design of diazabicyclooctane inhibitors of penicillin-binding proteins from Gram-negative bacteria to overcome multiple mechanisms of resistance, including ß-lactamase enzymes, stringent response and outer membrane permeation. Diazabicyclooctane inhibitors retain activity in the presence of ß-lactamases, the primary resistance mechanism associated with ß-lactam therapy in Gram-negative bacteria2,3. Although the target spectrum of an initial lead was successfully re-engineered to gain in vivo efficacy, its ability to permeate across bacterial outer membranes was insufficient for further development. Notably, the features that enhanced target potency were found to preclude compound uptake. An improved optimization strategy leveraged porin permeation properties concomitant with biochemical potency in the lead-optimization stage. This resulted in ETX0462, which has potent in vitro and in vivo activity against Pseudomonas aeruginosa plus all other Gram-negative ESKAPE pathogens, Stenotrophomonas maltophilia and biothreat pathogens. These attributes, along with a favourable preclinical safety profile, hold promise for the successful clinical development of the first novel Gram-negative chemotype to treat life-threatening antibiotic-resistant infections in more than 25 years.


Asunto(s)
Antibacterianos/farmacología , Diseño de Fármacos , Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas/efectos de los fármacos , Animales , Antibacterianos/química , Compuestos Aza/química , Compuestos Aza/farmacología , Ciclooctanos/química , Ciclooctanos/farmacología , Femenino , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Proteínas de Unión a las Penicilinas/antagonistas & inhibidores , Pseudomonas aeruginosa/efectos de los fármacos , beta-Lactamasas
3.
Chem Commun (Camb) ; 55(99): 14926-14929, 2019 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-31769767

RESUMEN

A technically simple approach for rapid, high-yielding and site-selective bioconjugation has been developed for both in vitro and cellular applications. This method involves the generation of maleimido-phosphonium ylides via 4-nitrophenol catalysis under physiological conditions followed by their Wittig reactions with aldehyde-appended biomolecules.


Asunto(s)
Aldehídos/química , Catálisis , Dicroismo Circular , Maleimidas/química , Estructura Molecular , Proteínas/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
4.
ACS Infect Dis ; 5(1): 49-54, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30489063

RESUMEN

The susceptibility of small molecules to Gram-negative bacterial efflux is typically evaluated using an antibacterial activity-based efflux ratio, which is computed as the ratio of the antibacterial activity for a wild-type strain and its isogenic efflux mutant (typically lacking genes encoding major efflux pumps). The magnitude of the ratio is often used as an efflux index. However, early in drug discovery, hits with suboptimal physicochemical properties often lack whole cell inhibition against wild-type strains, which makes efflux ratios indeterminable. To address this gap, we developed an assay to titrate levels of total efflux by varying the TolC expression using an arabinose-inducible promoter (pBAD) in an Escherichia coli Δ tolC strain. We provide a proof of concept for the assay using sets of related compounds from two antibiotic classes and show that the TolC titration provides a sensitive method for rank ordering compounds with respect to their efflux susceptibility.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/análisis , Proteínas de Escherichia coli/análisis , Escherichia coli/química , Proteínas de Transporte de Membrana/análisis , Arabinosa/química , Proteínas de la Membrana Bacteriana Externa/genética , Descubrimiento de Drogas , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Transporte de Membrana/genética , Pruebas de Sensibilidad Microbiana , Regiones Promotoras Genéticas , Prueba de Estudio Conceptual
5.
Res Microbiol ; 170(2): 112-116, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30468763

RESUMEN

The major Gram-negative gated efflux channel TolC has been extensively characterized in Escherichia coli but there is minimal information about Klebsiella pneumoniae TolC. Using an arabinose-inducible plasmid-based expression system, we show that the K. pneumoniae TolC complements the efflux defect in an E. coli K-12 ΔtolC strain, restoring wild-type levels of resistance towards most antibiotics suggesting that it can interact with the E. coli AcrB efflux pump. We characterize the efflux properties of K. pneumoniae TolC using an orthogonal whole cell-based assay and quantify the extrusion of environment-sensitive fluorescent probes and contrast the findings with the E. coli ortholog.


Asunto(s)
Antibacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de Escherichia coli , Proteínas de Transporte de Membrana/genética
6.
ACS Infect Dis ; 4(9): 1336-1345, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-29961312

RESUMEN

A general method for determining bacterial uptake of compounds independent of antibacterial activity would be a valuable tool in antibacterial drug discovery. LC-MS/MS assays have been described, but it has not been shown whether the data can be used directly to inform medicinal chemistry. We describe the evaluation of an LC-MS/MS assay measuring association of compounds with bacteria, using a set of over a hundred compounds (inhibitors of NAD-dependent DNA ligase, LigA) for which in vitro potency and antibacterial activity had been determined. All compounds were active against an efflux-deficient strain of Escherichia coli with reduced LigA activity ( E. coli ligA251 Δ tolC). Testing a single compound concentration and incubation time, we found that, for equipotent compounds, LC-MS/MS values were not predictive of antibacterial activity. This indicates that measured bacteria-associated compound was not necessarily exposed to the target enzyme. Our data suggest that, while exclusion from bacteria is a major reason for poor antibacterial activity of potent compounds, the distribution of compound within the bacterial cell may also be a problem. The relative importance of these factors is likely to vary from one chemical series to another. Our observations provide directions for further study of this difficult issue.


Asunto(s)
Antibacterianos/metabolismo , Cromatografía Liquida/métodos , Escherichia coli/química , Escherichia coli/metabolismo , Espectrometría de Masas en Tándem/métodos , Antibacterianos/análisis , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos
7.
ACS Infect Dis ; 4(5): 686-695, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29485271

RESUMEN

Our limited understanding of the molecular basis for compound entry into and efflux out of Gram-negative bacteria is now recognized as a key bottleneck for the rational discovery of novel antibacterial compounds. Traditional, large-scale biochemical or target-agnostic phenotypic antibacterial screening efforts have, as a result, not been very fruitful. A main driver of this knowledge gap has been the historical lack of predictive cellular assays, tools, and models that provide structure-activity relationships to inform optimization of compound accumulation. A variety of recent approaches has recently been described to address this conundrum. This Perspective explores these approaches and considers ways in which their integration could successfully redirect antibacterial drug discovery efforts.


Asunto(s)
Antibacterianos , Descubrimiento de Drogas , Antibacterianos/química , Antibacterianos/farmacología , Relación Dosis-Respuesta a Droga , Bacterias Gramnegativas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Permeabilidad , Relación Estructura-Actividad
8.
ACS Infect Dis ; 4(3): 373-381, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29260856

RESUMEN

OmpAAb is a conserved, abundantly expressed outer membrane porin in Acinetobacter baumannii whose presumed role in antibiotic permeation has not been clearly demonstrated. In this report, we use a titratable heterologous expression system to express OmpAAb in isolation and demonstrate selective passage of small molecule antibiotics through OmpAAb. ETX2514, a recently discovered broad-spectrum ß-lactamase inhibitor, in combination with sulbactam, is currently in clinical testing for the treatment of drug-resistant A. baumannii infections. We demonstrate that ETX2514 permeates OmpAAb and potentiates the activity of sulbactam in an OmpAAb-dependent manner. In addition, we show that small modifications in the structure of ETX2514 differentially affect its passage through OmpAAb, revealing unique structure-porin-permeation relationships. Finally, we confirm the contribution of OmpAAb to bacterial fitness using a murine thigh model of A. baumannii infection. These results, combined with the high sequence homology of OmpA across Acinetobacter spp., suggest that optimization of antibiotic entry through OmpAAb may prove to be a feasible medicinal chemistry design strategy for future antibacterial discovery efforts.


Asunto(s)
Acinetobacter baumannii/enzimología , Acinetobacter baumannii/metabolismo , Antibacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Porinas/metabolismo , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/fisiología , Animales , Compuestos de Azabiciclo/metabolismo , Transporte Biológico , Modelos Animales de Enfermedad , Aptitud Genética , Ratones , Relación Estructura-Actividad , Sulbactam/metabolismo
9.
ACS Infect Dis ; 3(4): 310-319, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28157293

RESUMEN

The global emergence of antibiotic resistance, especially in Gram-negative bacteria, is an urgent threat to public health. Discovery of novel classes of antibiotics with activity against these pathogens has been impeded by a fundamental lack of understanding of the molecular drivers underlying small molecule uptake. Although it is well-known that outer membrane porins represent the main route of entry for small, hydrophilic molecules across the Gram-negative cell envelope, the structure-permeation relationship for porin passage has yet to be defined. To address this knowledge gap, we developed a sensitive and specific whole-cell approach in Escherichia coli called titrable outer membrane permeability assay system (TOMAS). We used TOMAS to characterize the structure porin-permeation relationships of a set of novel carbapenem analogues through the Pseudomonas aeruginosa porin OprD. Our results show that small structural modifications, especially the number and nature of charges and their position, have dramatic effects on the ability of these molecules to permeate cells through OprD. This is the first demonstration of a defined relationship between specific molecular changes in a substrate and permeation through an isolated porin. Understanding the molecular mechanisms that impact antibiotic transit through porins should provide valuable insights to antibacterial medicinal chemistry and may ultimately allow for the rational design of porin-mediated uptake of small molecules into Gram-negative bacteria.


Asunto(s)
Carbapenémicos/química , Porinas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Carbapenémicos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Porinas/genética , Pseudomonas aeruginosa/metabolismo , Relación Estructura-Actividad
10.
Res Microbiol ; 166(6): 516-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26117599

RESUMEN

Resistance-Nodulation-Division (RND) family pumps AcrB and MexB are the major efflux routes in Escherichia coli and Pseudomonas aeruginosa respectively. Fluorescent environment-sensitive dyes provide a means to study efflux pump function in live bacterial cells in real-time. Recently, we demonstrated the utility of this approach using the dye Nile Red to quantify AcrB-mediated efflux and measured the ability of antibiotics and other efflux pump substrates to compete with efflux of Nile Red, independent of antibacterial activity. Here, we extend this method to P. aeruginosa and describe a novel application that permits the comparison and rank-ordering of bacterial strains by their inherent efflux potential. We show that glucose and l-malate re-energize Nile Red efflux in P. aeruginosa, and we highlight differences in the glucose dependence and kinetics of efflux between P. aeruginosa and E. coli. We quantify the differences in efflux among a set of P. aeruginosa laboratory strains, which include PAO1, the hyper-sensitive strain ATCC 35151 and its parent, ATCC 12055. Efflux of Nile Red in P. aeruginosa is mediated by MexAB-OprM and is slower than in E. coli. In conclusion, we describe an efflux measurement tool for use in antibacterial drug discovery and basic research on P. aeruginosa efflux pumps.


Asunto(s)
Proteínas Bacterianas/metabolismo , Descubrimiento de Drogas/métodos , Genes MDR , Oxazinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Antibacterianos/metabolismo , Colorantes Fluorescentes/metabolismo , Glucosa/metabolismo , Malatos/metabolismo
11.
Antimicrob Agents Chemother ; 59(4): 2388-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25645845

RESUMEN

A direct assay of efflux by Escherichia coli AcrAB-TolC and related multidrug pumps would have great value in discovery of new Gram-negative antibiotics. The current understanding of how efflux is affected by the chemical structure and physical properties of molecules is extremely limited, derived from antibacterial data for compounds that inhibit growth of wild-type E. coli. We adapted a previously described fluorescent efflux assay to a 96-well microplate format that measured the ability of test compounds to compete for efflux with Nile Red (an environment-sensitive fluor), independent of antibacterial activity. We show that Nile Red and the lipid-sensitive probe DiBAC4-(3) [bis-(1,3-dibutylbarbituric acid)-trimethine oxonol] can quantify efflux competition in E. coli. We extend the previous findings that the tetracyclines compete with Nile Red and show that DiBAC4-(3) competes with macrolides. The extent of the competition shows a modest correlation with the effect of the acrB deletion on MICs within the compound sets for both dyes. Crystallographic studies identified at least two substrate binding sites in AcrB, the proximal and distal pockets. High-molecular-mass substrates bound the proximal pocket, while low-mass substrates occupied the distal pocket. As DiBAC4-(3) competes with macrolides but not with Nile Red, we propose that DiBAC4-(3) binds the proximal pocket and Nile Red likely binds the distal site. In conclusion, competition with fluorescent probes can be used to study the efflux process for diverse chemical structures and may provide information as to the site of binding and, in some cases, enable rank-ordering a series of related compounds by efflux.


Asunto(s)
Antibacterianos/farmacología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Algoritmos , Sitios de Unión , Unión Competitiva , Escherichia coli/química , Proteínas de Escherichia coli/química , Colorantes Fluorescentes , Pruebas de Sensibilidad Microbiana , Peso Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Oxazinas
12.
Mol Microbiol ; 66(1): 1-13, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17880421

RESUMEN

We characterized two sucrose-metabolizing systems -sus and scr- and describe their roles in the physiology and virulence of Streptococcus pneumoniae in murine models of carriage and pneumonia. The sus and scr systems are regulated by LacI family repressors SusR and ScrR respectively. SusR regulates an adjacent ABC transporter (susT1/susT2/susX) and sucrose-6-phosphate (S-6-P) hydrolase (susH). ScrR controls an adjacent PTS transporter (scrT), fructokinase (scrK) and second S-6-P hydrolase (scrH). sus and scr play niche-specific roles in virulence. The susH and sus locus mutants are attenuated in the lung, but dispensable in nasopharyngeal carriage. Conversely, the scrH and scr locus mutants, while dispensable in the lung, are attenuated for nasopharyngeal colonization. The scrH/susH double mutant is more attenuated than scrH in the nasopharynx, indicating SusH can substitute in this niche. Both systems are sucrose-inducible, with ScrH being the major in vitro hydrolase. The scrH/susH mutant does not grow on sucrose indicating that sus and scr are the only sucrose-metabolizing systems in S. pneumoniae. We propose a model describing hierarchical regulation of the scr and sus systems by the putative inducer, S-6-P. The transport and metabolism of sucrose or a related disaccharide thus contributes to S. pneumoniae colonization and disease.


Asunto(s)
Redes y Vías Metabólicas , Streptococcus pneumoniae/metabolismo , Sacarosa/metabolismo , Animales , Portador Sano/microbiología , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Masculino , Ratones , Modelos Biológicos , Nasofaringe/microbiología , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crecimiento & desarrollo , Virulencia , beta-Fructofuranosidasa/genética , beta-Fructofuranosidasa/fisiología
13.
J Bacteriol ; 187(24): 8340-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16321938

RESUMEN

We characterized the role of catabolite control protein A (ccpA) in the physiology and virulence of Streptococcus pneumoniae. S. pneumoniae has a large percentage of its genome devoted to sugar uptake and metabolism, and therefore, regulation of these processes is likely to be crucial for fitness in the nasopharynx and may play a role during invasive disease. In many bacteria, carbon catabolite repression (CCR) is central to such regulation, influencing hierarchical sugar utilization and growth rates. CcpA is the major transcriptional regulator in CCR in several gram-positive bacteria. We show that CcpA functions in CCR of lactose-inducible beta-galactosidase activity in S. pneumoniae. CCR of maltose-inducible alpha-glucosidase, raffinose-inducible alpha-galactosidase, and cellobiose-inducible beta-glucosidase is unaffected in the ccpA strain, suggesting that other regulators, possibly redundant with CcpA, control these systems. The ccpA strain is severely attenuated for nasopharyngeal colonization and lung infection in the mouse, establishing its role in fitness on these mucosal surfaces. Comparison of the cell wall fraction of the ccpA and wild-type strains shows that CcpA regulates many proteins in this compartment that are involved in central and intermediary metabolism, a subset of which are required for survival and multiplication in vivo. Both in vitro and in vivo defects were complemented by providing ccpA in trans. Our results demonstrate that CcpA, though not a global regulator of CCR in S. pneumoniae, is required for colonization of the nasopharynx and survival and multiplication in the lung.


Asunto(s)
Proteínas Bacterianas/fisiología , Metabolismo de los Hidratos de Carbono , Proteínas de Unión al ADN/fisiología , Proteínas Represoras/fisiología , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad , Factores de Virulencia , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Portador Sano/microbiología , Pared Celular/química , Recuento de Colonia Microbiana , Proteínas de Unión al ADN/genética , Electroforesis en Gel Bidimensional , Regulación Bacteriana de la Expresión Génica , Prueba de Complementación Genética , Lactosa/metabolismo , Ratones , Mutagénesis Insercional , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Neumonía Neumocócica/microbiología , ARN Bacteriano/análisis , ARN Mensajero/análisis , Proteínas Represoras/genética , Streptococcus pneumoniae/genética , alfa-Galactosidasa/análisis , alfa-Glucosidasas/análisis , beta-Galactosidasa/análisis , beta-Glucosidasa/análisis
14.
Biochem J ; 376(Pt 1): 245-52, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12882645

RESUMEN

The L3 loop is an important feature of the OmpF porin structure, contributing to both channel size and electrostatic properties. Colicins A and N, spermine, and antibiotics that use OmpF to penetrate the cell, were used to investigate the structure-function relationships of L3. Spermine was found to protect efficiently cells expressing wild-type OmpF from colicin action. Among other solutes, sugars had minor effects on colicin A activity, whereas competitions between colicin A and antibiotic fluxes were observed. Among the antibiotics tested, cefepime appeared the most efficient. Escherichia coli cells expressing various OmpF proteins mutated in the eyelet were tested for their susceptibility to colicin A, and resistant strains were found only among L3 mutants. Mutations at residues 119 and 120 were the most effective at conferring resistance to colicin A, probably due to epitope structure alteration, as revealed by a specific antipeptide. More detailed information was obtained on mutants D113A and D121A, by focusing on the kinetics of colicin A and colicin N activities through measurements of potassium efflux. D113 appeared to play an essential role for colicin A activity, whereas colicin N activity was more dependent on D121 than on D113.


Asunto(s)
Cefalosporinas/metabolismo , Colicinas/metabolismo , Porinas/química , Porinas/metabolismo , Espermina/metabolismo , Antibacterianos , Unión Competitiva , Transporte Biológico , Western Blotting , Colicinas/antagonistas & inhibidores , Colicinas/farmacología , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Cinética , Modelos Moleculares , Mutación , Porinas/genética , Espermina/farmacología , Relación Estructura-Actividad
15.
Nature ; 419(6908): 715-8, 2002 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12384697

RESUMEN

An unexpected finding emerging from large-scale genome analyses is that prokaryotes express ion channels belonging to molecular families long studied in neurons. Bacteria and archaea are now known to carry genes for potassium channels of the voltage-gated, inward rectifier and calcium-activated classes, ClC-type chloride channels, an ionotropic glutamate receptor and a sodium channel. For two potassium channels and a chloride channel, these homologues have provided a means to direct structure determination. And yet the purposes of these ion channels in bacteria are unknown. Strong conservation of functionally important sequences from bacteria to vertebrates, and of structure itself, suggests that prokaryotes use ion channels in roles more adaptive than providing high-quality protein to structural biologists. Here we show that Escherichia coli uses chloride channels of the widespread ClC family in the extreme acid resistance response. We propose that the channels function as an electrical shunt for an outwardly directed virtual proton pump that is linked to amino acid decarboxylation.


Asunto(s)
Canales de Cloruro/metabolismo , Escherichia coli/metabolismo , Ácidos/metabolismo , Ácidos/farmacología , Arginina/metabolismo , Canales de Cloruro/genética , Cloruros/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen , Genes Bacterianos/genética , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Transporte Iónico , Liposomas/metabolismo , Fenotipo , Bombas de Protones/genética , Bombas de Protones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...