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.
mSystems ; 4(6)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31771976

RESUMEN

Interactions between the gut microbiome and immunoglobulin A (IgA) in the gut during infancy are important for future health. IgM and IgG are also present in the gut; however, their interactions with the microbiome in the developing infant remain to be characterized. Using stool samples sampled 15 times in infancy from 32 healthy subjects at 4 locations in 3 countries, we characterized patterns of microbiome development in relation to fecal levels of IgA, IgG, and IgM. For 8 infants from a single location, we used fluorescence-activated cell sorting of microbial cells from stool by Ig-coating status over 18 months. We used 16S rRNA gene profiling on full and sorted microbiomes to assess patterns of antibody coating in relation to age and other factors. All antibodies decreased in concentration with age but were augmented by breastmilk feeding regardless of infant age. Levels of IgA correlated with relative abundances of operational taxonomic units (OTUs) belonging to the Bifidobacteria and Enterobacteriaceae, which dominated the early microbiome, and IgG levels correlated with Haemophilus The diversity of Ig-coated microbiota was influenced by breastfeeding and age. IgA and IgM coated the same microbiota, which reflected the overall diversity of the microbiome, while IgG targeted a different subset. Blautia generally evaded antibody coating, while members of the Bifidobacteria and Enterobacteriaceae were high in IgA/M. IgA/M displayed similar dynamics, generally coating the microbiome proportionally, and were influenced by breastfeeding status. IgG only coated a small fraction of the commensal microbiota and differed from the proportion targeted by IgA and IgM.IMPORTANCE Antibodies are secreted into the gut and attach to roughly half of the trillions of bacterial cells present. When babies are born, the breastmilk supplies these antibodies until the baby's own immune system takes over this task after a few weeks. The vast majority of these antibodies are IgA, but two other types, IgG and IgM, are also present in the gut. Here, we ask if these three different antibody types target different types of bacteria in the infant gut as the infant develops from birth to 18 months old and how patterns of antibody coating of bacteria change with age. In this study of healthy infant samples over time, we found that IgA and IgM coat the same bacteria, which are generally representative of the diversity present, with a few exceptions that were more or less antibody coated than expected. IgG coated a separate suite of bacteria. These results provide a better understanding of how these antibodies interact with the developing infant gut microbiome.

2.
Environ Int ; 112: 279-286, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29316517

RESUMEN

Evolution has provided environmental bacteria with a plethora of genes that give resistance to antibiotic compounds. Under anthropogenic selection pressures, some of these genes are believed to be recruited over time into pathogens by horizontal gene transfer. River sediment polluted with fluoroquinolones and other drugs discharged from bulk drug production in India constitute an environment with unprecedented, long-term antibiotic selection pressures. It is therefore plausible that previously unknown resistance genes have evolved and/or are promoted here. In order to search for novel resistance genes, we therefore analyzed such river sediments by a functional metagenomics approach. DNA fragments providing resistance to different antibiotics in E. coli were sequenced using Sanger and PacBio RSII platforms. We recaptured the majority of known antibiotic resistance genes previously identified by open shot-gun metagenomics sequencing of the same samples. In addition, seven novel resistance gene candidates (six beta-lactamases and one amikacin resistance gene) were identified. Two class A beta-lactamases, blaRSA1 and blaRSA2, were phylogenetically close to clinically important ESBLs like blaGES, blaBEL and blaL2, and were further characterized for their substrate spectra. The blaRSA1 protein, encoded as an integron gene cassette, efficiently hydrolysed penicillins, first generation cephalosporins and cefotaxime, while blaRSA2 was an inducible class A beta-lactamase, capable of hydrolyzing carbapenems albeit with limited efficiency, similar to the L2 beta-lactamase from Stenotrophomonas maltophilia. All detected novel genes were associated with plasmid mobilization proteins, integrons, and/or other resistance genes, suggesting a potential for mobility. This study provides insight into a resistome shaped by an exceptionally strong and long-term antibiotic selection pressure. An improved knowledge of mobilized resistance factors in the external environment may make us better prepared for the resistance challenges that we may face in clinics in the future.


Asunto(s)
Carbapenémicos/metabolismo , Sedimentos Geológicos/microbiología , Metagenoma , Contaminantes Químicos del Agua/análisis , Resistencia betalactámica , beta-Lactamasas/genética , Antibacterianos/farmacología , Industria Farmacéutica , Residuos Industriales/efectos adversos , Metagenoma/efectos de los fármacos , Metagenoma/genética , Metagenómica , Ríos/microbiología , Resistencia betalactámica/efectos de los fármacos , Resistencia betalactámica/genética
3.
Gastric Cancer ; 20(1): 116-125, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26782287

RESUMEN

BACKGROUND: An increase of regulatory T cells, defined as CD25high- and/or FOXP3+-expressing CD4+ T cells, within tumors has been reported in several studies. Tregs promote tumor growth by modulating the antitumor immune response, mainly through inhibition of T-cell-mediated tumor cell killing: this has been suggested to be dependent on IL-10 and/or TGF-ß. In stomach cancer, the mechanisms behind the accumulation of Tregs in tumor tissue has not been fully elucidated, and neither has Treg gene expression in situ. MATERIALS AND METHODS: Stomach tissue from gastric cancer patients undergoing gastric resection was analyzed using flow cytometry and cell sorting, followed by RT-PCR. RESULTS: We observed that stomach CD4+ FOXP3+ T cells proliferated to a higher degree than CD4+ FOXP3- T cells, which may contribute to Treg accumulation in the mucosa. By analyzing DNA methylation, we demonstrated that both proliferating and nonproliferating FOXP3+ T cells exhibited complete demethylation of the FOXP3 gene, indicating a stable FOXP3 expression in both cell populations. Furthermore, analysis of T-cell populations isolated directly from the tumor and tumor-free mucosa demonstrated that CD4+ CD25high T cells have a higher IL-10/IFN-γ gene expression ratio but express lower levels of TGF-ß than CD4+ CD25low/- T cells. CONCLUSION: We demonstrate strong proliferation among regulatory CD4+ FOXP3+ CD25high T cells in the gastric cancer mucosa. These local Treg express a suppressive cytokine profile characterized by high IL-10 and low TGF-ß and IFN-γ production.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Mucosa Gástrica/inmunología , Interleucina-10/genética , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/genética , Anciano , Anciano de 80 o más Años , Apoptosis , Biomarcadores de Tumor/genética , Metilación de ADN , Femenino , Citometría de Flujo , Estudios de Seguimiento , Factores de Transcripción Forkhead/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/sangre , Neoplasias Gástricas/genética , Células Tumorales Cultivadas
4.
Appl Environ Microbiol ; 82(4): 1050-9, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26637602

RESUMEN

Metagenomes derived from environmental microbiota encode a vast diversity of protein homologs. How this diversity impacts protein function can be explored through selection assays aimed to optimize function. While artificially generated gene sequence pools are typically used in selection assays, their usage may be limited because of technical or ethical reasons. Here, we investigate an alternative strategy, the use of soil microbial DNA as a starting point. We demonstrate this approach by optimizing the function of a widely occurring soil bacterial enzyme, 1-aminocyclopropane-1-carboxylate (ACC) deaminase. We identified a specific ACC deaminase domain region (ACCD-DR) that, when PCR amplified from the soil, produced a variant pool that we could swap into functional plasmids carrying ACC deaminase-encoding genes. Functional clones of ACC deaminase were selected for in a competition assay based on their capacity to provide nitrogen to Escherichia coli in vitro. The most successful ACCD-DR variants were identified after multiple rounds of selection by sequence analysis. We observed that previously identified essential active-site residues were fixed in the original unselected library and that additional residues went to fixation after selection. We identified a divergent essential residue whose presence hints at the possible use of alternative substrates and a cluster of neutral residues that did not influence ACCD performance. Using an artificial ACCD-DR variant library generated by DNA oligomer synthesis, we validated the same fixation patterns. Our study demonstrates that soil metagenomes are useful starting pools of protein-coding-gene diversity that can be utilized for protein optimization and functional characterization when synthetic libraries are not appropriate.


Asunto(s)
Alelos , Liasas de Carbono-Carbono/aislamiento & purificación , Liasas de Carbono-Carbono/metabolismo , Pruebas Genéticas/métodos , Metagenómica/métodos , Rizosfera , Microbiología del Suelo , Liasas de Carbono-Carbono/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Nitrógeno/metabolismo
5.
J Infect Dis ; 212(9): 1480-90, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25883383

RESUMEN

BACKGROUND: Staphylococcus epidermidis causes late-onset sepsis in preterm infants. Staphylococcus epidermidis activates host responses in part via Toll-like receptor 2 (TLR2). Epidemiologic studies link bacteremia and neonatal brain injury, but direct evidence is lacking. METHODS: Wild-type and TLR2-deficient (TLR2-/-) mice were injected intravenously with S. epidermidis at postnatal day 1 prior to measuring plasma and brain cytokine and chemokine levels, bacterial clearance, brain caspase-3 activation, white/gray matter volume, and innate transcriptome. RESULTS: Staphylococcus epidermidis bacteremia spontaneously resolved over 24 hours without detectable bacteria in the cerebrospinal fluid (CSF). TLR2-/- mice demonstrated delayed S. epidermidis clearance from blood, spleen, and liver. Staphylococcus epidermidis increased the white blood cell count in the CSF, increased interleukin 6, interleukin 12p40, CCL2, and CXCL1 concentrations in plasma; increased the CCL2 concentration in the brain; and caused rapid (within 6 hours) TLR2-dependent brain activation of caspase-3 and TLR2-independent white matter injury. CONCLUSIONS: Staphylococcus epidermidis bacteremia, in the absence of bacterial entry into the CSF, impairs neonatal brain development. Staphylococcus epidermidis bacteremia induced both TLR2-dependent and -independent brain injury, with the latter occurring in the absence of TLR2, a condition associated with an increased bacterial burden. Our study indicates that the consequences of transient bacteremia in early life may be more severe than commonly appreciated, and our findings may inform novel approaches to reduce bacteremia-associated brain injury.


Asunto(s)
Bacteriemia/patología , Lesiones Encefálicas/microbiología , Staphylococcus epidermidis/aislamiento & purificación , Receptor Toll-Like 2/metabolismo , Animales , Animales Recién Nacidos , Caspasa 3/genética , Caspasa 3/metabolismo , Quimiocina CCL2/sangre , Quimiocina CXCL1/sangre , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Subunidad p40 de la Interleucina-12/sangre , Interleucina-6/sangre , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/microbiología , Receptor Toll-Like 2/genética , Regulación hacia Arriba
6.
Environ Sci Technol ; 48(14): 7825-32, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24988042

RESUMEN

There is increasing concern that environmental antibiotic pollution promotes transfer of resistance genes to the human microbiota. Here, fluoroquinolone-polluted river sediment, well water, irrigated farmland, and human fecal flora of local villagers within a pharmaceutical industrial region in India were analyzed for quinolone resistance (qnr) genes by quantitative PCR. Similar samples from Indian villages farther away from industrial areas, as well as fecal samples from Swedish study participants and river sediment from Sweden, were included for comparison. Fluoroquinolones were detected by MS/MS in well water and soil from all villages located within three km from industrially polluted waterways. Quinolone resistance genes were detected in 42% of well water, 7% of soil samples and in 100% and 18% of Indian and Swedish river sediments, respectively. High antibiotic concentrations in Indian sediment coincided with high abundances of qnr, whereas lower fluoroquinolone levels in well water and soil did not. We could not find support for an enrichment of qnr in fecal samples from people living in the fluoroquinolone-contaminated villages. However, as qnr was detected in 91% of all Indian fecal samples (24% of the Swedish) it suggests that the spread of qnr between people is currently a dominating transmission route.


Asunto(s)
Contaminación Ambiental/análisis , Heces/microbiología , Fluoroquinolonas/análisis , Genes Bacterianos/genética , Sedimentos Geológicos/química , Residuos Industriales/análisis , Contaminantes del Suelo/análisis , Contaminantes Químicos del Agua/análisis , Adolescente , Adulto , Anciano , Antibacterianos/análisis , Niño , Preescolar , ADN Ribosómico/genética , Femenino , Dosificación de Gen , Sedimentos Geológicos/microbiología , Humanos , India , Masculino , Persona de Mediana Edad , Ríos/química , Población Rural , Suelo/química , Adulto Joven
7.
PLoS One ; 9(4): e93943, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24714675

RESUMEN

Both Th1 and Th17 cells are important components of the immune response to Helicobacter pylori (Hp) in adults, but less is known about T cell responses to Hp during early childhood, when the infection is often acquired. We investigated Th1 and Th17 type responses to Hp in adults, children and infants in Bangladesh, where Hp is highly endemic. IL-17 and IFN-γ mRNA levels in gastric biopsies from Hp-infected Bangladeshi adults were analyzed and compared to levels in infected and uninfected Swedish controls. Since biopsies could not be collected from infants and children, cytokine responses in Bangladeshi infants (6-12 months), children (3-5 years) and adults (>19 years) were instead compared by stimulating peripheral blood mononuclear cells (PBMCs) with a Hp membrane preparation (MP) and analyzing culture supernatants by ELISA and cytometric bead array. We found significantly higher expression of IL-17 and IFN-γ mRNA in gastric mucosa of Hp-infected Bangladeshi and Swedish adults compared to uninfected Swedish controls. PBMCs from all age groups produced IL-17 and IFN-γ after MP stimulation, but little Th2 cytokines. IL-17 and IFN-γ were primarily produced by CD4+ T cells, since CD4+ T cell depleted PBMCs produced reduced amounts of these cytokines. Infant cells produced significantly more IL-17, but similar levels of IFN-γ, compared to adult cells after MP stimulation. In contrast, polyclonal stimulation induced lower levels IL-17 and IFN-γ in infant compared to adult PBMCs and CD4+ T cells. The strong IL-17 production in infants after MP stimulation was paralleled by significantly higher production of the IL-17 promoting cytokine IL-1ß from infant compared to adult PBMCs and monocytes. In conclusion, these results show that T cells can produce high levels of IL-17 and IFN-γ in response to Hp from an early age and indicate a potential role for IL-1ß in promoting Th17 responses to Hp during infancy.


Asunto(s)
Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Células TH1/metabolismo , Células Th17/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Bangladesh , Preescolar , Femenino , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Humanos , Lactante , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Masculino , Persona de Mediana Edad , Suecia , Células TH1/patología , Células Th17/patología , Adulto Joven
8.
Cell Host Microbe ; 14(5): 571-81, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24237702

RESUMEN

Gut mucosal barrier breakdown and inflammation have been associated with high levels of flagellin, the principal bacterial flagellar protein. Although several gut commensals can produce flagella, flagellin levels are low in the healthy gut, suggesting the existence of control mechanisms. We find that mice lacking the flagellin receptor Toll-like receptor 5 (TLR5) exhibit a profound loss of flagellin-specific immunoglobulins (Igs) despite higher total Ig levels in the gut. Ribotyping of IgA-coated cecal microbiota showed Proteobacteria evading antibody coating in the TLR5(-/-) gut. A diversity of microbiome members overexpressed flagellar genes in the TLR5(-/-) host. Proteobacteria and Firmicutes penetrated small intestinal villi, and flagellated bacteria breached the colonic mucosal barrier. In vitro, flagellin-specific Ig inhibited bacterial motility and downregulated flagellar gene expression. Thus, innate-immunity-directed development of flagellin-specific adaptive immune responses can modulate the microbiome's production of flagella in a three-way interaction that helps to maintain mucosal barrier integrity and homeostasis.


Asunto(s)
Flagelina/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Inmunidad Mucosa , Locomoción , Microbiota/inmunología , Inmunidad Adaptativa , Animales , Flagelos/inmunología , Inmunidad Innata , Inmunoglobulina A/inmunología , Ratones , Ratones Noqueados , Receptor Toll-Like 5/deficiencia , Receptor Toll-Like 5/inmunología
9.
PLoS One ; 6(2): e17038, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21359229

RESUMEN

The high and sometimes inappropriate use of antibiotics has accelerated the development of antibiotic resistance, creating a major challenge for the sustainable treatment of infections world-wide. Bacterial communities often respond to antibiotic selection pressure by acquiring resistance genes, i.e. mobile genetic elements that can be shared horizontally between species. Environmental microbial communities maintain diverse collections of resistance genes, which can be mobilized into pathogenic bacteria. Recently, exceptional environmental releases of antibiotics have been documented, but the effects on the promotion of resistance genes and the potential for horizontal gene transfer have yet received limited attention. In this study, we have used culture-independent shotgun metagenomics to investigate microbial communities in river sediments exposed to waste water from the production of antibiotics in India. Our analysis identified very high levels of several classes of resistance genes as well as elements for horizontal gene transfer, including integrons, transposons and plasmids. In addition, two abundant previously uncharacterized resistance plasmids were identified. The results suggest that antibiotic contamination plays a role in the promotion of resistance genes and their mobilization from environmental microbes to other species and eventually to human pathogens. The entire life-cycle of antibiotic substances, both before, under and after usage, should therefore be considered to fully evaluate their role in the promotion of resistance.


Asunto(s)
Antibacterianos/efectos adversos , Biota , Farmacorresistencia Bacteriana , Transferencia de Gen Horizontal/efectos de los fármacos , Sedimentos Geológicos/microbiología , Análisis de Secuencia de ADN/métodos , Contaminantes Químicos del Agua/efectos adversos , Bacterias/efectos de los fármacos , Bacterias/genética , Elementos Transponibles de ADN/fisiología , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Transferencia de Gen Horizontal/fisiología , Sedimentos Geológicos/química , Humanos , Ríos/química , Microbiología del Agua
10.
Helicobacter ; 14(4): 237-47, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19674127

RESUMEN

BACKGROUND: Helicobacter pylori is one of the most prevalent human bacterial pathogens; however, its transmission pathways remain unknown. New infections of H. pylori during outbreaks of gastroenteritis have been suggested previously, and to explore this transmission route further H. pylori was quantified in vomitus and diarrheal stool of patients suffering from acute gastroenteritis in Dhaka, Bangladesh. MATERIALS AND METHODS: Vomitus and stool samples from 28 patients seeking care at the International Centre for Diarrhoeal Disease Research hospital were analyzed for presence of H. pylori and other pathogens using quantitative culturing, real-time polymerase chain reaction (PCR), and H. pylori stool antigen test. Bacterial gene expression was analyzed using reverse transcriptase real-time PCR. RESULTS: The results of real-time PCR show that 23 (88%) of the 26 vomitus samples and 17 (74%) of the 23 stool samples were H. pylori positive, while stool antigen test show that 14 (67%) of the 21 stool samples were H. pylori positive. H. pylori could not be isolated by culture. Analysis using quantitative culture and real-time PCR to detect Vibrio cholerae showed strong correlation between these methods, and validating real-time PCR. Analysis of H. pylori virulence gene transcription in vomitus, diarrheal stool, antral and duodenal biopsy specimens, and in vitro cultures showed that cagA, flaA, and ureA were highly transcribed in vomitus, biopsy specimens, and cultures, whereas hpaA and vacA were expressed at lower levels. No H. pylori gene expression was detected in diarrheal stool. CONCLUSIONS: We conclude that high numbers of transcriptionally active H. pylori are shed in vomitus, which indicates that new infections may be disseminated through vomiting.


Asunto(s)
Gastroenteritis/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/aislamiento & purificación , Vómitos/microbiología , Enfermedad Aguda/epidemiología , Adolescente , Adulto , Anciano , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Bangladesh/epidemiología , Niño , Heces/microbiología , Femenino , Gastroenteritis/epidemiología , Infecciones por Helicobacter/epidemiología , Humanos , Hidroliasas/genética , Masculino , Persona de Mediana Edad , Oxidorreductasas/genética , Vómitos/epidemiología , Adulto Joven
11.
Appl Environ Microbiol ; 75(10): 3039-44, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19304824

RESUMEN

The main transmission pathway of Helicobacter pylori has not been determined, but several reports have described detection of H. pylori DNA in drinking and environmental water, suggesting that H. pylori may be waterborne. To address this possibility, we developed, tested, and optimized two complementary H. pylori-specific real-time PCR assays for quantification of H. pylori DNA in water. The minimum detection level of the assays including collection procedures and DNA extraction was shown to be approximately 250 H. pylori genomes per water sample. Using our assays, we then analyzed samples of drinking and environmental water (n = 75) and natural water biofilms (n = 21) from a high-endemicity area in Bangladesh. We could not identify H. pylori DNA in any of the samples, even though other pathogenic bacteria have been found previously in the same water samples by using the same methodology. A series of control experiments were performed to ensure that the negative results were not falsely caused by PCR inhibition, nonspecific assays, degradation of template DNA, or low detection sensitivity. Our results suggest that it is unlikely that the predominant transmission route of H. pylori in this area is waterborne.


Asunto(s)
ADN Bacteriano/aislamiento & purificación , Helicobacter pylori/aislamiento & purificación , Reacción en Cadena de la Polimerasa/métodos , Microbiología del Agua , Bangladesh , ADN Bacteriano/genética , Helicobacter pylori/genética , Sensibilidad y Especificidad
12.
Microb Pathog ; 44(3): 246-54, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18037262

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) cause diarrhoea by adhesion to human enterocytes by one or more colonization factors (CFs) and secretion of heat-labile (LT) and/or heat-stable (ST) enterotoxins. Expression of coli surface antigen 6 (CS6) on the bacterial surface, usually associated with ETEC strains that produce ST alone or in combination with LT, is rarely found in strains expressing only LT. However, a number of LT-only strains which are genotypically positive but phenotypically negative for CS6 have been identified. In this study, eight such strains from India and Guinea-Bissau belonging to different clones were analysed. The CS6 operon cssABCD was transcribed but protein analyses suggested that the structural subunits CssA and CssB of CS6 were absent in the periplasm. Most strains contained truncating mutations within the periplasmic chaperone-encoding gene cssC and protein modelling indicated that this severely affected the substrate-binding capacity of the chaperone. A single-nucleotide polymorphism (SNP) (A-->T) in the 5'-untranslated region of cssC distinguished the eight strains from ETEC strains that do express CS6 on the surface and may be a potential marker for ETEC strains containing phenotypically silent cssABCD. The study emphasizes the importance of using both genotypic and phenotypic methods in epidemiological studies of ETEC, e.g. for vaccine development.


Asunto(s)
Antígenos Bacterianos/metabolismo , Escherichia coli Enterotoxigénica/metabolismo , Proteínas de Escherichia coli/metabolismo , Mutación/genética , Antígenos Bacterianos/análisis , Antígenos Bacterianos/genética , Escherichia coli Enterotoxigénica/genética , Escherichia coli Enterotoxigénica/inmunología , Enterotoxinas , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/análisis , Proteínas de Escherichia coli/genética , Chaperonas Moleculares/genética , Operón , Proteínas Periplasmáticas/metabolismo , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...