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.
Methods Protoc ; 6(2)2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37104024

RESUMEN

Outer membrane vesicles (OMVs) are lipid structures containing various biomolecules in their native environment and are spontaneously shed by gram-negative bacteria. OMVs perform several biological functions critical to both bacterial physiology and pathogenicity. Scientific research on OMV function and biogenesis requires a standardized and robust method of isolating these vesicles from bacterial cultures that reliably provide high-purity OMVs. Herein, we describe an optimized protocol to isolate OMVs from overnight cultures of three different strains of nontypeable Haemophilus influenzae (NTHi) for use in different downstream applications. Involving mainly differential centrifugation of the culture supernatant, the procedure described is relatively simple, efficient, and generates high-quality OMV preparations from each strain tested with sufficient yields, while preserving the native outer membrane composition.

2.
J Biol Chem ; 292(42): 17290-17301, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-28842504

RESUMEN

Clostridium difficile is a major nosocomial pathogen that produces two exotoxins, TcdA and TcdB, with TcdB thought to be the primary determinant in human disease. TcdA and TcdB are large, multidomain proteins, each harboring a cytotoxic glucosyltransferase domain that is delivered into the cytosol from endosomes via a translocation domain after receptor-mediated endocytosis of toxins from the cell surface. Although there are currently no known host cell receptors for TcdA, three cell-surface receptors for TcdB have been identified: CSPG4, NECTIN3, and FZD1/2/7. The sites on TcdB that mediate binding to each receptor are not defined. Furthermore, it is not known whether the combined repetitive oligopeptide (CROP) domain is involved in or required for receptor binding. Here, in a screen designed to identify sites in TcdB that are essential for target cell intoxication, we identified a region at the junction of the translocation and the CROP domains that is implicated in CSPG4 binding. Using a series of C-terminal truncations, we show that the CSPG4-binding site on TcdB extends into the CROP domain, requiring three short repeats for binding and for full toxicity on CSPG4-expressing cells. Consistent with the location of the CSPG4-binding site on TcdB, we show that the anti-TcdB antibody bezlotoxumab, which binds partially within the first three short repeats, prevents CSPG4 binding to TcdB. In addition to establishing the binding region for CSPG4, this work ascribes for the first time a role in TcdB CROPs in receptor binding and further clarifies the relative roles of host receptors in TcdB pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Clostridioides difficile/enzimología , Glucosiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/genética , Anticuerpos ampliamente neutralizantes , Células CHO , Células CACO-2 , Chlorocebus aethiops , Proteoglicanos Tipo Condroitín Sulfato/genética , Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Cricetinae , Cricetulus , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/genética , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Dominios Proteicos
3.
J Mol Biol ; 429(7): 1030-1044, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28232034

RESUMEN

The exotoxins toxin A (TcdA) and toxin B (TcdB) are produced by the bacterial pathogen Clostridium difficile and are responsible for the pathology associated with C. difficile infection (CDI). The antitoxin antibodies actoxumab and bezlotoxumab bind to and neutralize TcdA and TcdB, respectively. Bezlotoxumab was recently approved by the FDA for reducing the recurrence of CDI. We have previously shown that a single molecule of bezlotoxumab binds to two distinct epitopes within the TcdB combined repetitive oligopeptide (CROP) domain, preventing toxin binding to host cells. In this study, we characterize the binding of actoxumab to TcdA and examine its mechanism of toxin neutralization. Using a combination of approaches including a number of biophysical techniques, we show that there are two distinct actoxumab binding sites within the CROP domain of TcdA centered on identical amino acid sequences at residues 2162-2189 and 2410-2437. Actoxumab binding caused the aggregation of TcdA especially at higher antibody:toxin concentration ratios. Actoxumab prevented the association of TcdA with target cells demonstrating that actoxumab neutralizes toxin activity by inhibiting the first step of the intoxication cascade. This mechanism of neutralization is similar to that observed with bezlotoxumab and TcdB. Comparisons of the putative TcdA epitope sequences across several C. difficile ribotypes and homologous repeat sequences within TcdA suggest a structural basis for observed differences in actoxumab binding and/or neutralization potency. These data provide a mechanistic basis for the protective effects of the antibody in vitro and in vivo, including in various preclinical models of CDI.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/metabolismo , Toxinas Bacterianas/antagonistas & inhibidores , Enterotoxinas/antagonistas & inhibidores , Epítopos/metabolismo , Sitios de Unión , Anticuerpos ampliamente neutralizantes , Agregado de Proteínas , Unión Proteica
4.
Antimicrob Agents Chemother ; 60(11): 6471-6482, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27527088

RESUMEN

Clostridium difficile causes infections of the colon in susceptible patients. Specifically, gut dysbiosis induced by treatment with broad-spectrum antibiotics facilitates germination of ingested C. difficile spores, expansion of vegetative cells, and production of symptom-causing toxins TcdA and TcdB. The current standard of care for C. difficile infections (CDI) consists of administration of antibiotics such as vancomycin that target the bacterium but also perpetuate gut dysbiosis, often leading to disease recurrence. The monoclonal antitoxin antibodies actoxumab (anti-TcdA) and bezlotoxumab (anti-TcdB) are currently in development for the prevention of recurrent CDI. In this study, the effects of vancomycin or actoxumab/bezlotoxumab treatment on progression and resolution of CDI were assessed in mice and hamsters. Rodent models of CDI are characterized by an early severe phase of symptomatic disease, associated with high rates of morbidity and mortality; high intestinal C. difficile burden; and a disrupted intestinal microbiota. This is followed in surviving animals by gradual recovery of the gut microbiota, associated with clearance of C. difficile and resolution of disease symptoms over time. Treatment with vancomycin prevents disease initially by inhibiting outgrowth of C. difficile but also delays microbiota recovery, leading to disease relapse following discontinuation of therapy. In contrast, actoxumab/bezlotoxumab treatment does not impact the C. difficile burden but rather prevents the appearance of toxin-dependent symptoms during the early severe phase of disease, effectively preventing disease until the microbiota (the body's natural defense against C. difficile) has fully recovered. These data provide insight into the mechanism of recurrence following vancomycin administration and into the mechanism of recurrence prevention observed clinically with actoxumab/bezlotoxumab.


Asunto(s)
Antibacterianos/efectos adversos , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Antitoxinas/farmacología , Infecciones por Clostridium/tratamiento farmacológico , Vancomicina/efectos adversos , Animales , Antibacterianos/administración & dosificación , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/biosíntesis , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/biosíntesis , Anticuerpos ampliamente neutralizantes , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/mortalidad , Convalecencia , Cricetulus , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enterotoxinas/antagonistas & inhibidores , Enterotoxinas/biosíntesis , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia , Vancomicina/administración & dosificación
5.
Antimicrob Agents Chemother ; 59(2): 1052-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25451052

RESUMEN

Clostridium difficile infections (CDIs) are the leading cause of hospital-acquired infectious diarrhea and primarily involve two exotoxins, TcdA and TcdB. Actoxumab and bezlotoxumab are human monoclonal antibodies that neutralize the cytotoxic/cytopathic effects of TcdA and TcdB, respectively. In a phase II clinical study, the actoxumab-bezlotoxumab combination reduced the rate of CDI recurrence in patients who were also treated with standard-of-care antibiotics. However, it is not known whether the antibody combination will be effective against a broad range of C. difficile strains. As a first step toward addressing this, we tested the ability of actoxumab and bezlotoxumab to neutralize the activities of toxins from a number of clinically relevant and geographically diverse strains of C. difficile. Neutralization potencies, as measured in a cell growth/survival assay with purified toxins from various C. difficile strains, correlated well with antibody/toxin binding affinities. Actoxumab and bezlotoxumab neutralized toxins from culture supernatants of all clinical isolates tested, including multiple isolates of the BI/NAP1/027 and BK/NAP7/078 strains, at antibody concentrations well below plasma levels observed in humans. We compared the bezlotoxumab epitopes in the TcdB receptor binding domain across known TcdB sequences and found that key substitutions within the bezlotoxumab epitopes correlated with the relative differences in potencies of bezlotoxumab against TcdB of some strains, including ribotypes 027 and 078. Combined with in vitro neutralization data, epitope modeling will enhance our ability to predict the coverage of new and emerging strains by actoxumab-bezlotoxumab in the clinic.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Clostridioides difficile/efectos de los fármacos , Proteínas Bacterianas/genética , Línea Celular , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Epítopos/inmunología , Femenino , Humanos , Masculino
6.
Infect Immun ; 83(2): 822-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25486992

RESUMEN

Clostridium difficile infection (CDI) represents the most prevalent cause of antibiotic-associated gastrointestinal infections in health care facilities in the developed world. Disease symptoms are caused by the two homologous exotoxins, TcdA and TcdB. Standard therapy for CDI involves administration of antibiotics that are associated with a high rate of disease recurrence, highlighting the need for novel treatment paradigms that target the toxins rather than the organism itself. A combination of human monoclonal antibodies, actoxumab and bezlotoxumab, directed against TcdA and TcdB, respectively, has been shown to decrease the rate of recurrence in patients treated with standard-of-care antibiotics. However, the exact mechanism of antibody-mediated protection is poorly understood. In this study, we show that the antitoxin antibodies are protective in multiple murine models of CDI, including systemic and local (gut) toxin challenge models, as well as primary and recurrent models of infection in mice. Systemically administered actoxumab-bezlotoxumab prevents both the damage to the gut wall and the inflammatory response, which are associated with C. difficile in these models, including in mice challenged with a strain of the hypervirulent ribotype 027. Furthermore, mutant antibodies (N297Q) that do not bind to Fcγ receptors provide a level of protection similar to that of wild-type antibodies, demonstrating that the mechanism of protection is through direct neutralization of the toxins and does not involve host effector functions. These data provide a mechanistic basis for the prevention of recurrent disease observed in CDI patients in clinical trials.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Antitoxinas/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/prevención & control , Enterotoxinas/inmunología , Animales , Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Antitoxinas/uso terapéutico , Chlorocebus aethiops , Modelos Animales de Enfermedad , Enterocolitis Seudomembranosa/inmunología , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores de IgG/inmunología , Recurrencia , Células Vero
7.
J Biol Chem ; 289(26): 18008-21, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24821719

RESUMEN

The symptoms of Clostridium difficile infections are caused by two exotoxins, TcdA and TcdB, which target host colonocytes by binding to unknown cell surface receptors, at least in part via their combined repetitive oligopeptide (CROP) domains. A combination of the anti-TcdA antibody actoxumab and the anti-TcdB antibody bezlotoxumab is currently under development for the prevention of recurrent C. difficile infections. We demonstrate here through various biophysical approaches that bezlotoxumab binds to specific regions within the N-terminal half of the TcdB CROP domain. Based on this information, we solved the x-ray structure of the N-terminal half of the TcdB CROP domain bound to Fab fragments of bezlotoxumab. The structure reveals that the TcdB CROP domain adopts a ß-solenoid fold consisting of long and short repeats and that bezlotoxumab binds to two homologous sites within the CROP domain, partially occluding two of the four putative carbohydrate binding pockets located in TcdB. We also show that bezlotoxumab neutralizes TcdB by blocking binding of TcdB to mammalian cells. Overall, our data are consistent with a model wherein a single molecule of bezlotoxumab neutralizes TcdB by binding via its two Fab regions to two epitopes within the N-terminal half of the TcdB CROP domain, partially blocking the carbohydrate binding pockets of the toxin and preventing toxin binding to host cells.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Clostridioides difficile/inmunología , Epítopos/inmunología , Secuencia de Aminoácidos , Anticuerpos Antibacterianos/química , Anticuerpos Monoclonales , Anticuerpos Neutralizantes/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Sitios de Unión , Anticuerpos ampliamente neutralizantes , Clostridioides difficile/química , Clostridioides difficile/genética , Cristalografía por Rayos X , Mapeo Epitopo , Epítopos/química , Epítopos/genética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
8.
Science ; 304(5678): 1805-7, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15205533

RESUMEN

Salmonella enterica, the cause of food poisoning and typhoid fever, induces actin cytoskeleton rearrangements and membrane ruffling to gain access into nonphagocytic cells, where it can replicate and avoid innate immune defenses. Here, we found that SopB, a phosphoinositide phosphatase that is delivered into host cells by a type III secretion system, was essential for the establishment of Salmonella's intracellular replicative niche. SopB mediated the formation of spacious phagosomes following bacterial entry and was responsible for maintaining high levels of phosphatidylinositol-three-phosphate [PtdIns(3)P] in the membrane of the bacteria-containing vacuoles. Absence of SopB caused a significant defect in the maturation of the Salmonella-containing vacuole and impaired bacterial intracellular growth.


Asunto(s)
Proteínas Bacterianas/metabolismo , Vesículas Citoplasmáticas/microbiología , Mucosa Intestinal/microbiología , Fagosomas/microbiología , Fosfatidilinositoles/metabolismo , Salmonella typhimurium/metabolismo , Antígenos CD/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Células Epiteliales/microbiología , Eliminación de Gen , Islas Genómicas , Humanos , Mucosa Intestinal/citología , Proteínas de Membrana de los Lisosomas , Microscopía por Video , Mutación , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Vacuolas/metabolismo , Vacuolas/microbiología , Vacuolas/ultraestructura
9.
J Cell Biol ; 163(5): 1123-31, 2003 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-14662750

RESUMEN

Salmonella enterica, the causative agent of food poisoning and typhoid fever, induces programmed cell death in macrophages, a process found to be dependent on a type III protein secretion system, and SipB, a protein with membrane fusion activity that is delivered into host cells by this system. When expressed in cultured cells, SipB caused the formation of and localized to unusual multimembrane structures. These structures resembled autophagosomes and contained both mitochondrial and endoplasmic reticulum markers. A mutant form of SipB devoid of membrane fusion activity localized to mitochondria, but did not induce the formation of membrane structures. Upon Salmonella infection of macrophages, SipB was found in mitochondria, which appeared swollen and devoid of christae. Salmonella-infected macrophages exhibited marked accumulation of autophagic vesicles. We propose that Salmonella, through the action of SipB, kills macrophages by disrupting mitochondria, thereby inducing autophagy and cell death.


Asunto(s)
Autofagia/fisiología , Proteínas Bacterianas/metabolismo , Muerte Celular , Macrófagos/fisiología , Salmonella typhimurium/metabolismo , Animales , Proteínas Bacterianas/genética , Células COS , Caspasa 1/genética , Caspasa 1/metabolismo , Retículo Endoplásmico/metabolismo , Macrófagos/citología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Salmonella typhimurium/patogenicidad
11.
Cell ; 110(2): 191-202, 2002 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-12150927

RESUMEN

Toll-like receptors (TLRs) detect microorganisms and protect multicellular organisms from infection. TLRs transduce their signals through MyD88 and the serine/threonine kinase IRAK. The IRAK family consists of two active kinases, IRAK and IRAK-4, and two inactive kinases, IRAK-2 and IRAK-M. IRAK-M expression is restricted to monocytes/macrophages, whereas other IRAKs are ubiquitous. We show here that IRAK-M is induced upon TLR stimulation and negatively regulates TLR signaling. IRAK-M prevented dissociation of IRAK and IRAK-4 from MyD88 and formation of IRAK-TRAF6 complexes. IRAK-M(-/-) cells exhibited increased cytokine production upon TLR/IL-1 stimulation and bacterial challenge, and IRAK-M(-/-) mice showed increased inflammatory responses to bacterial infection. Endotoxin tolerance, a protection mechanism against endotoxin shock, was significantly reduced in IRAK-M(-/-) cells. Thus, IRAK-M regulates TLR signaling and innate immune homeostasis.


Asunto(s)
Proteínas de Drosophila , Glicoproteínas de Membrana/inmunología , Proteínas Quinasas/inmunología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos de Diferenciación/biosíntesis , Secuencia de Bases , Línea Celular Transformada , Células Cultivadas , Clonación Molecular , ADN Complementario , Escherichia coli/inmunología , Femenino , Humanos , Interleucina-1/farmacología , Quinasas Asociadas a Receptores de Interleucina-1 , Interleucina-12/biosíntesis , Interleucina-6/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Listeria monocytogenes , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/inmunología , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide , FN-kappa B/inmunología , Biosíntesis de Proteínas , Proteínas Quinasas/genética , Receptores Inmunológicos/biosíntesis , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología , Salmonella typhimurium/inmunología , Factor 6 Asociado a Receptor de TNF , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos
12.
Nature ; 416(6877): 194-9, 2002 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-11894098

RESUMEN

The immune system consists of two evolutionarily different but closely related responses, innate immunity and adaptive immunity. Each of these responses has characteristic receptors-Toll-like receptors (TLRs) for innate immunity and antigen-specific receptors for adaptive immunity. Here we show that the caspase recruitment domain (CARD)-containing serine/threonine kinase Rip2 (also known as RICK, CARDIAK, CCK and Ripk2) transduces signals from receptors of both immune responses. Rip2 was recruited to TLR2 signalling complexes after ligand stimulation. Moreover, cytokine production in Rip2-deficient cells was reduced on stimulation of TLRs with lipopolysaccharide, peptidoglycan and double-stranded RNA, but not with bacterial DNA, indicating that Rip2 is downstream of TLR2/3/4 but not TLR9. Rip2-deficient cells were also hyporesponsive to signalling through interleukin (IL)-1 and IL-18 receptors, and deficient for signalling through Nod proteins-molecules also implicated in the innate immune response. Furthermore, Rip2-deficient T cells showed severely reduced NF-kappaB activation, IL-2 production and proliferation on T-cell-receptor (TCR) engagement, and impaired differentiation to T-helper subtype 1 (TH1) cells, indicating that Rip2 is required for optimal TCR signalling and T-cell differentiation. Rip2 is therefore a signal transducer and integrator of signals for both the innate and adaptive immune systems.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de Drosophila , Inmunidad Innata/inmunología , Péptidos y Proteínas de Señalización Intracelular , Listeriosis/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Proteínas Portadoras/metabolismo , Diferenciación Celular , Células Cultivadas , Eliminación de Gen , Interleucinas/biosíntesis , Interleucinas/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/metabolismo , Listeriosis/patología , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1 , Proteína Adaptadora de Señalización NOD2 , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Citocinas/biosíntesis , Receptores de Interleucina/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor Toll-Like 2 , Receptores Toll-Like , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...