Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(44): 22307-22313, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31611409

RESUMEN

Aggregatibacter actinomycetemcomitans is associated with aggressive periodontitis resulting in premature tooth loss in adolescents. Tooth adherence and biofilm persistence are prerequisites for survival in the oral domain. Here, using a rhesus monkey model, 16S rRNA sequencing, and weighted network analysis, we assessed colonization of A. actinomycetemcomitans variants and ascertained microbial interactions in biofilm communities. Variants in A. actinomycetemcomitans leukotoxin (ltx) were created, labeled, inoculated, and compared with their progenitor strain for in vivo colonization. Samples of tooth-related plaque were assessed for colonization at baseline and after debridement and inoculation of labeled strains. Null, minimal, and hyper-Ltx-producing strains were created and assessed for hydroxyapatite binding and biofilm formation in vitro. Ltx-hyperproducing strains colonized with greater prevalence and at higher levels than wild type or ltx mutants (P = 0.05). Indigenous and inoculated A. actinomycetemcomitans strains that attached were associated with lactate-producing species (i.e., Leptotrichia, Abiotrophia, and Streptoccocci). A. actinomycetemcomitans was found at 0.13% of the total flora at baseline and at 0.05% 4 wk after inoculation. In vivo data were supported by in vitro results. We conclude that hyper-Ltx production affords these strains with an attachment advantage providing a foothold for competition with members of the indigenous microbiota. Increased attachment can be linked to ltx gene expression and up-regulation of adherence-associated genes. Growth of attached A. actinomycetemcomitans in vivo was enhanced by lactate availability due to consorting species. These associations provide A. actinomycetemcomitans with the constituents required for its colonization and survival in the complex and competitive oral environment.


Asunto(s)
Aggregatibacter actinomycetemcomitans/patogenicidad , Boca/microbiología , Periodontitis/microbiología , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/metabolismo , Aggregatibacter actinomycetemcomitans/fisiología , Animales , Adhesión Bacteriana/efectos de los fármacos , Biopelículas , Durapatita/farmacología , Exotoxinas/genética , Exotoxinas/metabolismo , Ácido Láctico/metabolismo , Macaca mulatta , Masculino , Microbiota
2.
J Infect Dis ; 224(8): 1345-1356, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34387310

RESUMEN

BACKGROUND: We studied risk factors, antibodies, and symptoms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in a diverse, ambulatory population. METHODS: A prospective cohort (n = 831) previously undiagnosed with SARS-CoV-2 infection underwent serial testing (SARS-CoV-2 polymerase chain reaction, immunoglobulin G [IgG]) for 6 months. RESULTS: Ninety-three participants (11.2%) tested SARS-CoV-2-positive: 14 (15.1%) asymptomatic, 24 (25.8%) severely symptomatic. Healthcare workers (n = 548) were more likely to become infected (14.2% vs 5.3%; adjusted odds ratio, 2.1; 95% confidence interval, 1.4-3.3) and severely symptomatic (29.5% vs 6.7%). IgG antibodies were detected after 79% of asymptomatic infections, 89% with mild-moderate symptoms, and 96% with severe symptoms. IgG trajectories after asymptomatic infections (slow increases) differed from symptomatic infections (early peaks within 2 months). Most participants (92%) had persistent IgG responses (median 171 days). In multivariable models, IgG titers were positively associated with symptom severity, certain comorbidities, and hospital work. Dyspnea and neurologic changes (including altered smell/taste) lasted ≥ 120 days in ≥ 10% of affected participants. Prolonged symptoms (frequently more severe) corresponded to higher antibody levels. CONCLUSIONS: In a prospective, ethnically diverse cohort, symptom severity correlated with the magnitude and trajectory of IgG production. Symptoms frequently persisted for many months after infection.Clinical Trials Registration. NCT04336215.


Asunto(s)
Anticuerpos Antivirales/sangre , COVID-19/diagnóstico , Inmunoglobulina G/sangre , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Adulto , Anticuerpos Antivirales/inmunología , Infecciones Asintomáticas/epidemiología , COVID-19/sangre , COVID-19/epidemiología , COVID-19/transmisión , Comorbilidad , Femenino , Humanos , Inmunoglobulina G/inmunología , Incidencia , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , SARS-CoV-2/inmunología , Adulto Joven
3.
Antimicrob Agents Chemother ; 56(5): 2397-407, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22330917

RESUMEN

The in vitro antibacterial effects of diallyl sulfide (DAS) against the Gram-negative periodontopathogen Aggregatibacter actinomycetemcomitans, the key etiologic agent of the severe form of localized aggressive periodontitis and other nonoral infections, were studied. A. actinomycetemcomitans was treated with garlic extract, allicin, or DAS, and the anti-A. actinomycetemcomitans effects of the treatment were evaluated. Garlic extract, allicin, and DAS significantly inhibited the growth of A. actinomycetemcomitans (greater than 3 log; P < 0.01) compared to control cells. Heat inactivation of the garlic extracts significantly reduced the protein concentration; however, the antimicrobial effect was retained. Purified proteins from garlic extract did not exhibit antimicrobial activity. Allicin lost all its antimicrobial effect when it was subjected to heat treatment, whereas DAS demonstrated an antimicrobial effect similar to that of the garlic extract, suggesting that the antimicrobial activity of garlic extract is mainly due to DAS. An A. actinomycetemcomitans biofilm-killing assay performed with DAS showed a significant reduction in biofilm cell numbers, as evidenced by both confocal microscopy and culture. Scanning electron microscopy (SEM) analysis of DAS-treated A. actinomycetemcomitans biofilms showed alterations of colony architecture indicating severe stress. Flow cytometry analysis of OBA9 cells did not demonstrate apoptosis or cell cycle arrest at therapeutic concentrations of DAS (0.01 and 0.1 µg/ml). DAS-treated A. actinomycetemcomitans cells demonstrated complete inhibition of glutathione (GSH) S-transferase (GST) activity. However, OBA9 cells, when exposed to DAS at similar concentrations, showed no significant differences in GST activity, suggesting that DAS-induced GST inhibition might be involved in A. actinomycetemcomitans cell death. These findings demonstrate that DAS exhibits significant antibacterial activity against A. actinomycetemcomitans and that this property might be utilized for exploring its therapeutic potential in treatment of A. actinomycetemcomitans-associated oral and nonoral infections.


Asunto(s)
Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Compuestos Alílicos/farmacología , Antibacterianos/farmacología , Ajo/química , Extractos Vegetales/química , Sulfuros/farmacología , Infecciones por Actinobacillus/tratamiento farmacológico , Infecciones por Actinobacillus/microbiología , Aggregatibacter actinomycetemcomitans/enzimología , Aggregatibacter actinomycetemcomitans/ultraestructura , Periodontitis Agresiva/tratamiento farmacológico , Periodontitis Agresiva/microbiología , Compuestos Alílicos/aislamiento & purificación , Antibacterianos/aislamiento & purificación , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Supervivencia Celular/efectos de los fármacos , Disulfuros , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Encía/citología , Encía/efectos de los fármacos , Glutatión Transferasa/antagonistas & inhibidores , Glutatión Transferasa/metabolismo , Calor , Humanos , Viabilidad Microbiana/efectos de los fármacos , Microscopía Electrónica de Rastreo , Extractos Vegetales/farmacología , Sulfuros/aislamiento & purificación , Ácidos Sulfínicos/aislamiento & purificación , Ácidos Sulfínicos/farmacología
4.
mBio ; 12(2)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758084

RESUMEN

Like the bacterial residents of the human gut, it is likely that many of the species in the human oral microbiota have evolved to better occupy and persist in their niche. Aggregatibacter actinomycetemcomitans (Aa) is both a common colonizer of the oral cavity and has been implicated in the pathogenesis of periodontal disease. Here, we present a whole-genome phylogenetic analysis of Aa isolates from humans and nonhuman primates that revealed an ancient origin for this species and a long history of association with the Catarrhini, the lineage that includes Old World monkeys (OWM) and humans. Further genomic analysis showed a strong association with the presence of a short-chain fatty acid (SCFA) catabolism locus (atoRDAEB) in many human isolates that was absent in almost all nonhuman OWM isolates. We show that this locus was likely acquired through horizontal gene transfer. When grown under conditions that are similar to those at the subgingival site of periodontitis (anaerobic, SCFA replete), Aa strains with atoRDAEB formed robust biofilms and showed upregulation of genes involved in virulence, colonization, and immune evasion. Both an isogenic deletion mutant and nonhuman primate isolates lacking the ato locus failed to grow in a robust biofilm under these conditions, but grew well under the carbohydrate-rich conditions similar to those found above the gumline. We propose that the acquisition of the ato locus was a key evolutionary step allowing Aa to utilize SCFAs, adapt, and modulate subgingival disease.IMPORTANCE There has been considerable interest in the impact of short-chain fatty acids (SCFAs) on inflammatory effects related to the microbiome. Here, we present evidence that SCFAs may also be important in disease by providing an energy source or disease-associated cue for colonizing pathogens. We propose that SCFAs allow Aggregatibacter actinomycetemcomitans (Aa) to adapt to the subgingival anaerobic environment, which is the site of human periodontitis. Under anaerobic, SCFA-rich conditions, human-derived Aa strains that possess butyrate metabolism genes form strong biofilms and upregulate virulence genes. Our phylogenetic analysis highlights a long history of evolution of Aa with its primate hosts and suggests that the acquisition of butyrate metabolism genes may have been a critical step in allowing Aa to colonize a new niche and cause disease in humans. Overall, this study highlights the important role that horizontal gene transfer may play in microbial adaptation and the evolution of infectious disease.


Asunto(s)
Adaptación Fisiológica/genética , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/metabolismo , Butiratos/metabolismo , Transferencia de Gen Horizontal , Aggregatibacter actinomycetemcomitans/patogenicidad , Anaerobiosis , Biopelículas , Genoma Bacteriano , Factores de Tiempo
5.
Pathogens ; 9(3)2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32131551

RESUMEN

Aggregatibacter actinomycetemcomitans, the focus of this review, was initially proposed as a microbe directly related to a phenotypically distinct form of periodontitis called localized juvenile periodontitis. At the time, it seemed as if specific microbes were implicated as the cause of distinct forms of disease. Over the years, much has changed. The sense that specific microbes relate to distinct forms of disease has been challenged, as has the sense that distinct forms of periodontitis exist. This review consists of two components. The first part is presented as a detective story where we attempt to determine what role, if any, Aggregatibacter plays as a participant in disease. The second part describes landscape ecology in the context of how the host environment shapes the framework of local microbial dysbiosis. We then conjecture as to how the local host response may limit the damage caused by pathobionts. We propose that the host may overcome the constant barrage of a dysbiotic microbiota by confining it to a local tooth site. We conclude speculating that the host response can confine local damage by restricting bacteremic translocation of members of the oral microbiota to distant organs thus constraining morbidity and mortality of the host.

6.
Front Immunol ; 10: 728, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31040843

RESUMEN

Aggregatibacter actinomycetemcomitans (Aa) is a low-abundance Gram-negative oral pathobiont that is highly associated with a silent but aggressive orphan disease that results in periodontitis and tooth loss in adolescents of African heritage. For the most part Aa conducts its business by utilizing strategies allowing it to conceal itself below the radar of the host mucosal immune defense system. A great deal of misinformation has been conveyed with respect to Aa biology in health and disease. The purpose of this review is to present misconceptions about Aa and the strategies that it uses to colonize, survive, and evade the host. In the process Aa manages to undermine host mucosal defenses and contribute to disease initiation. This review will present clinical observational, molecular, and interventional studies that illustrate genetic, phenotypic, and biogeographical tactics that have been recently clarified and demonstrate how Aa survives and suppresses host mucosal defenses to take part in disease pathogenesis. At one point in time Aa was considered to be the causative agent of Localized Aggressive Periodontitis. Currently, it is most accurate to look at Aa as a community activist and necessary partner of a pathogenic consortium that suppresses the initial host response so as to encourage overgrowth of its partners. The data for Aa's activist role stems from molecular genetic studies complemented by experimental animal investigations that demonstrate how Aa establishes a habitat (housing), nutritional sustenance in that habitat (food), and biogeographical mobilization and/or relocation from its initial habitat (transportation). In this manner Aa can transfer to a protected but vulnerable domain (pocket or sulcus) where its community activism is most useful. Aa's "strategy" includes obtaining housing, food, and transportation at no cost to its partners challenging the economic theory that "there ain't no such thing as a free lunch." This "strategy" illustrates how co-evolution can promote Aa's survival, on one hand, and overgrowth of community members, on the other, which can result in local host dysbiosis and susceptibility to infection.


Asunto(s)
Aggregatibacter actinomycetemcomitans/patogenicidad , Periodontitis Agresiva/microbiología , Infecciones por Pasteurellaceae/microbiología , Adolescente , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/inmunología , Periodontitis Agresiva/etiología , Periodontitis Agresiva/inmunología , Animales , Biopelículas/crecimiento & desarrollo , Genes Bacterianos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Mucosa , Modelos Inmunológicos , Infecciones por Pasteurellaceae/etiología , Infecciones por Pasteurellaceae/inmunología
7.
Mol Oral Microbiol ; 32(5): 432-442, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28383798

RESUMEN

Experiments were designed to explore a prominent autoinducer-2 (AI-2) producing gene (luxS) related to colonization and survival of Aggregatibacter actinomycetemcomitans, a low abundance member of the indigenous flora, that forms a key component of the dysbiotic flora in localized aggressive periodontitis. The luxS gene was disrupted in a primate strain of A. actinomycetemcomitans before implantation into the oral cavity of Rhesus monkeys (Rh). The colonization efficiency of the luxS mutant (RhAa-VS4) was compared with the parental wild-type strain (RhAa3) (positive control) and a ltxA mutant (RhAa-VS2) (negative control). The in vivo results showed that the luxS mutation had minimal impact on A. actinomycetemcomitans colonization compared with the wild-type RhAa3 strain. In vitro studies revealed that there was a significant upregulation of attachment-related genes aae, apiA, and flp in the RhAa-VS4 strain compared with RhAa3. Biofilm forming ability was also significantly increased in the RhAa-VS4 strain compared with RhAa3, whereas the AI-2 signal was ablated. The exogenous addition of the AI-2 precursor dihydroxy pentanedione allowed the RhAa-VS4 strain to achieve RhAa3 biofilm levels. This is the first primate study to test the relevance of LuxS in vivo. In vitro assessment suggests that in vivo survival of the RhAa-VS4 strain was due to the production of signaling AI-2 molecules derived from other members of the flora as well as the upregulation of genes related to attachment and biofilm formation.


Asunto(s)
Adaptación Fisiológica/genética , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/fisiología , Proteínas Bacterianas/genética , Liasas de Carbono-Azufre/genética , Boca/microbiología , Mutación , Animales , Proteínas Bacterianas/metabolismo , Biopelículas , Liasas de Carbono-Azufre/metabolismo , Regulación Bacteriana de la Expresión Génica , Homoserina/análogos & derivados , Homoserina/genética , Homoserina/metabolismo , Lactonas/metabolismo , Macaca mulatta , Viabilidad Microbiana , Percepción de Quorum/genética
8.
Sci Rep ; 7(1): 1887, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507341

RESUMEN

Aggregatibacter actinomycetemcomitans leukotoxin (LtxA) is a major virulence factor that kills leukocytes permitting it's escape from host immune surveillance. A. actinomycetemcomitans strains can produce high or low levels of toxin. Genetic differences reside in the "so called JP2" ltxA promoter region. These hyper-leukotoxin producing strains with the 530 bp deletion have been studied in detail. However, regions contained within the 530 bp deletion that could be responsible for modulation of leukotoxin production have not been defined. Here, we report, for the first time, on regions within the 530 bp that are responsible for high-levels of ltxA expression. We constructed a deletion of 530 bps in a primate isolate of A. actinomycetemcomitans, which produced leukotoxin equivalent to the JP2 strain. We then constructed sequential deletions in regions that span the 530 bps. Results indicated that expression of the ltxA transcript was reduced by a potential transcriptional terminator in promoter region 298 to 397 with a ΔG = -7.9 kcal/mol. We also confirmed previous findings that transcriptional fusion between the orfX region and ltxC increased ltxA expression. In conclusion, we constructed a hyper-leukotoxin producing A. actinomycetemcomitans strain and identified a terminator located in the promoter region extending from 298-397 that alters ltxA expression.


Asunto(s)
Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/metabolismo , Exotoxinas/biosíntesis , Exotoxinas/genética , Regiones Promotoras Genéticas , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Secuencia de Bases , Exotoxinas/química , Regulación Bacteriana de la Expresión Génica , Humanos , Mutación , Conformación de Ácido Nucleico , Operón , Terminación de la Transcripción Genética , Transcripción Genética
9.
PLoS One ; 11(3): e0151361, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26977924

RESUMEN

Leukotoxin (Ltx) is a prominent virulence factor produced by Aggregatibacter actinomycetemcomitans, an oral microorganism highly associated with aggressive periodontitis. Ltx compromises host responsiveness by altering the viability of neutrophils, lymphocytes, and macrophages. Previously, we developed a Rhesus (Rh) monkey colonization model designed to determine the effect of virulence gene mutations on colonization of A. actinomycetemcomitans. Unexpectedly, an A. actinomycetemcomitans leukotoxin (ltxA) mutant (RhAa-VS2) failed to colonize in the Rh model. No previous literature suggested that Ltx was associated with A. actinomycetemcomitans binding to tooth surfaces. These results led us to explore the broad effects of the ltxA mutation in vitro. Results indicated that LtxA activity was completely abolished in RhAa-VS2 strain, while complementation significantly (P<0.0001) restored leukotoxicity compared to RhAa-VS2 strain. RT-PCR analysis of ltx gene expression ruled out polar effects. Furthermore, binding of RhAa-VS2 to salivary-coated hydroxyapatite (SHA) was significantly decreased (P<0.0001) compared to wild type RhAa3 strain. Real time RT-PCR analysis of the genes related to SHA binding in RhAa-VS2 showed that genes related to binding were downregulated [rcpA (P = 0.018), rcpB (P = 0.02), tadA (P = 0.002)] as compared to wild type RhAa3. RhAa-VS2 also exhibited decreased biofilm depth (P = 0.008) and exo-polysaccharide production (P<0.0001). Buccal epithelial cell (BEC) binding of RhAa-VS2 was unaffected. Complementation with ltxA restored binding to SHA (P<0.002) but had no effect on biofilm formation when compared to RhAa3. In conclusion, mutation of ltxA diminished hard tissue binding in vitro, which helps explain the previous in vivo failure of a ltxA knockout to colonize the Rh oral cavity. These results suggest that; 1) one specific gene knockout (in this case ltxA) could affect other seemingly unrelated genes (such as rcpA, rcpB tadA etc), and 2) some caution should be used when interpreting the effect attributed to targeted gene mutations when seen in a competitive in vivo environment.


Asunto(s)
Aggregatibacter actinomycetemcomitans/genética , Toxinas Bacterianas/genética , Exotoxinas/genética , Mutación , Factores de Virulencia/genética , Aggregatibacter actinomycetemcomitans/metabolismo , Aggregatibacter actinomycetemcomitans/patogenicidad , Animales , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Macaca mulatta , Factores de Virulencia/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-26213715

RESUMEN

Aggregatibacter actinomycetemcomitans (Aa) is a pathobiont and part of a consortium of bacteria that can lead to periodontitis in humans. Our aim was to develop a model for oral inoculation of labeled Aa into a suitable host in order to study Aa traits and ecological factors that either enhance or repress its persistence. Primate species were screened for Aa to select a host for colonization studies. Macaca mulatta (Rhesus/Rh) was selected. Rh Aa strains were isolated, subjected to sequencing and functional analysis for comparison to human strains. "Best" methods for microbial decontamination prior to inoculation were assessed. Three groups were studied; Group 1 (N=5) was inoculated with Aa Spectinomycin resistant (SpecR) Rh strain 4.35, Group 2 (N=5) inoculated with Aa SpecR human strain IDH 781, and Group 3 (N=5) the un-inoculated control. Repeated feeding with pancakes spiked with SpecRAa followed high dose oral inoculation. Cheek, tongue, and plaque samples collected at baseline 1, 2, 3, and 4 weeks after inoculation were plated on agar; 1) selective for Aa, 2) enriched for total counts, and 3) containing 50 µg/ml of Spec. Aa was identified by colonial morphology and DNA analysis. Rh and human Aa had > 93-98 % genome identity. Rh Aa attached to tissues better than IDH 781 in vitro (p < 0.05). SpecR IDH 781 was not recovered from any tissue at any time; whereas, RhSpecR 4.35 was detected in plaque, but never tongue or cheek, in all monkeys at all times (> 1 × 105 colonies/ml; p < 0.001). In conclusion, the primate model provides a useful platform for studying integration of Aa strains into a reduced but established oral habitat. Primate derived SpecRAa was consistently detected in plaque at all collection periods; however, human derived Aa was never detected. The model demonstrated both microbial as well as tissue specificity.

11.
Microbes Infect ; 16(9): 762-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25124544

RESUMEN

Streptococcus mutans is the primary agent of dental caries, which is often detected in transient bacteremia. Lactoferrin is a multifunctional glycoprotein showing antibacterial activities against several Streptococcus species. We reported here the prophylactic effect of human lactoferrin (hLF) in a lactoferrin knockout mouse (LFKO-/-) bacteremic model. The hLF treatment significantly cleared S. mutans from the blood and organs of bacteremic mice when compared to the non-hLF treated mice. Further, analysis of serum cytokines, spleen and liver cytokine mRNA levels revealed that hLF prophylaxis modulates their release differently when compared to the non-hLF treated group. C-reactive protein level (P = 0.003) also decreased following hLF prophylaxis in S. mutans induced bacteremic mice. Additional quantitative RT-PCR analysis revealed that hLF prophylaxis significantly decreased the expression level of IFN-γ, TNF-α, IL-1ß, IL-6, MPO and iNOS in spleen and liver. These results suggested that the hLF protects the host against S. mutans-induced experimental bacteremia.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriemia/prevención & control , Lactoferrina/farmacología , Infecciones Estreptocócicas/prevención & control , Streptococcus mutans/efectos de los fármacos , Animales , Bacteriemia/inmunología , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Factor Estimulante de Colonias de Granulocitos/genética , Humanos , Interferón gamma/sangre , Interferón gamma/genética , Interleucina-1beta/sangre , Interleucina-1beta/genética , Interleucina-3/genética , Interleucina-6/sangre , Interleucina-6/genética , Lactoferrina/genética , Hígado/efectos de los fármacos , Hígado/inmunología , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/sangre , Óxido Nítrico Sintasa de Tipo II/genética , Proteínas Recombinantes de Fusión/genética , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/ultraestructura , Infecciones Estreptocócicas/inmunología , Streptococcus mutans/crecimiento & desarrollo , Streptococcus mutans/patogenicidad , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA