Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38729159

RESUMEN

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Asunto(s)
Adyuvantes Inmunológicos , Linfocitos T CD8-positivos , Quitina , Proteínas de la Membrana , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta , Transducción de Señal , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Receptor de Interferón alfa y beta/genética , Ratones , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Humanos , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Femenino , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias/inmunología , Neoplasias/terapia
2.
Cell Rep Med ; 4(1): 100899, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36652908

RESUMEN

The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.


Asunto(s)
Inflamasomas , Nanopartículas , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Caspasas/metabolismo , Interleucina-1/metabolismo
3.
Sci Immunol ; 7(78): eade5728, 2022 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-36525507

RESUMEN

Interleukin-1 (IL-1) family cytokines are key barrier cytokines that are typically expressed as inactive, or partially active, precursors that require proteolysis within their amino termini for activation. IL-37 is an enigmatic member of the IL-1 family that has been proposed to be activated by caspase-1 and to exert anti-inflammatory activity through engagement of the IL-18R and SIGIRR. However, here we show that the longest IL-37 isoform, IL-37b, exhibits robust proinflammatory activity upon amino-terminal proteolysis by neutrophil elastase or cathepsin S. In sharp contrast, caspase-1 failed to process or activate IL-37 at concentrations that robustly activated its canonical substrate, IL-1ß. IL-37 and IL-36 exhibit high structural homology, and, consistent with this, a K53-truncated form of IL-37, mimicking the cathepsin S-processed form of this cytokine, was found to exert its proinflammatory effects via IL-36 receptor engagement and produced an inflammatory signature practically identical to IL-36. Administration of K53-truncated IL-37b intraperitoneally into wild-type mice also elicited an inflammatory response that was attenuated in IL-36R-/- animals. These data demonstrate that, in common with other IL-1 family members, mature IL-37 can also elicit proinflammatory effects upon processing by specific proteases.


Asunto(s)
Interleucina-1 , Péptido Hidrolasas , Receptores de Interleucina , Animales , Ratones , Caspasas , Catepsinas , Citocinas , Interleucina-1/metabolismo , Células Mieloides , Receptores de Interleucina/metabolismo
4.
Biomaterials ; 275: 120961, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34171753

RESUMEN

Chitosan is a cationic polysaccharide that has been evaluated as an adjuvant due to its biocompatible and biodegradable nature. The polysaccharide can enhance antibody responses and cell-mediated immunity following vaccination by injection or mucosal routes. However, the optimal polymer characteristics for activation of dendritic cells (DCs) and induction of antigen-specific cellular immune responses have not been resolved. Here, we demonstrate that only chitin-derived polymers with a high degree of deacetylation (DDA) enhance generation of mitochondrial reactive oxygen species (mtROS), leading to cGAS-STING mediated induction of type I IFN. Additionally, the capacity of the polymers to activate the NLRP3 inflammasome was strictly dependent on the degree and pattern of deacetylation and mtROS generation. Polymers with a DDA below 80% are poor adjuvants while a fully deacetylated polyglucosamine polymer is most effective as a vaccine adjuvant. Furthermore, this polyglucosamine polymer enhanced antigen-specific Th1 responses in a NLRP3 and STING-type I IFN-dependent manner. Overall these results indicate that the degree of chitin deacetylation, the acetylation pattern and its regulation of mitochondrial ROS are the key determinants of its immune enhancing effects.


Asunto(s)
Inflamasomas , Proteínas de la Membrana , Proteína con Dominio Pirina 3 de la Familia NLR , Quitina , Mitocondrias , Nucleotidiltransferasas , Polímeros , Especies Reactivas de Oxígeno
5.
Respir Res ; 22(1): 133, 2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33926483

RESUMEN

Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.


Asunto(s)
Inmunidad Adaptativa , Asma/inmunología , Enfermedades Transmisibles/inmunología , Inmunidad Innata , Pulmón/inmunología , Metales/inmunología , Microbiota , Estado Nutricional , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Animales , Asma/microbiología , Asma/fisiopatología , Asma/virología , Enfermedades Transmisibles/microbiología , Enfermedades Transmisibles/fisiopatología , Enfermedades Transmisibles/virología , Interacciones Huésped-Patógeno , Humanos , Pulmón/microbiología , Pulmón/fisiopatología , Pulmón/virología , Metales/metabolismo , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/virología
6.
Semin Immunol ; 56: 101542, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34973890

RESUMEN

The dogma that immunological memory is an exclusive trait of adaptive immunity has been recently challenged by studies showing that priming of innate cells can also result in modified long-term responsiveness to secondary stimuli, once the cells have returned to a non-activated state. This phenomenon is known as 'innate immune memory', 'trained immunity' or 'innate training'. While the main known triggers of trained immunity are microbial-derived molecules such as ß-glucan, endogenous particles such as oxidized low-density lipoprotein and monosodium urate crystals can also induce trained phenotypes in innate cells. Whether exogenous particles can induce trained immunity has been overlooked. Our exposure to particulates has dramatically increased in recent decades as a result of the broad medical use of particle-based drug carriers, theragnostics, adjuvants, prosthetics and an increase in environmental pollution. We recently showed that pristine graphene can induce trained immunity in macrophages, enhancing their inflammatory response to TLR agonists, proving that exogenous nanomaterials can affect the long-term response of innate cells. The consequences of trained immunity can be beneficial, for instance, enhancing protection against unrelated pathogens; however, they can also be deleterious if they enhance inflammatory disorders. Therefore, studying the ability of particulates and biomaterials to induce innate trained phenotypes in cells is warranted. Here we analyse the mechanisms whereby particles can induce trained immunity and discuss how physicochemical characteristics of particulates could influence the induction of innate memory. We review the implications of trained immunity in the context of particulate adjuvants, nanocarriers and nanovaccines and their potential applications in medicine. Finally, we reflect on the unanswered questions and the future of the field.


Asunto(s)
Inmunidad Innata , Nanopartículas , Inmunidad Adaptativa , Adyuvantes Inmunológicos , Humanos , Memoria Inmunológica , Macrófagos
7.
PLoS One ; 15(11): e0239713, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33211703

RESUMEN

Uncoupling proteins (UCPs) are members of the mitochondrial anion carrier superfamily that can mediate the transfer of protons into the mitochondrial matrix from the intermembrane space. We have previously reported UCP3 expression in thymocytes, mitochondria of total splenocytes and splenic lymphocytes. Here, we demonstrate that Ucp3 is expressed in peripheral naive CD4+ T cells at the mRNA level before being markedly downregulated following activation. Non-polarized, activated T cells (Th0 cells) from Ucp3-/- mice produced significantly more IL-2, had increased expression of CD25 and CD69 and were more proliferative than Ucp3+/+ Th0 cells. The altered IL-2 expression observed between T cells from Ucp3+/+ and Ucp3-/- mice may be a factor in determining differentiation into Th17 or induced regulatory (iTreg) cells. When compared to Ucp3+/+, CD4+ T cells from Ucp3-/- mice had increased FoxP3 expression under iTreg conditions. Conversely, Ucp3-/- CD4+ T cells produced a significantly lower concentration of IL-17A under Th17 cell-inducing conditions in vitro. These effects were mirrored in antigen-specific T cells from mice immunized with KLH and CT. Interestingly, the altered responses of Ucp3-/- T cells were partially reversed upon neutralisation of IL-2. Together, these data indicate that UCP3 acts to restrict the activation of naive T cells, acting as a rheostat to dampen signals following TCR and CD28 co-receptor ligation, thereby limiting early activation responses. The observation that Ucp3 ablation alters the Th17:Treg cell balance in vivo as well as in vitro suggests that UCP3 is a potential target for the treatment of Th17 cell-mediated autoimmune diseases.


Asunto(s)
Activación de Linfocitos/genética , Linfocitos T Reguladores/citología , Células Th17/citología , Proteína Desacopladora 3/genética , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular/genética , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Proteína Desacopladora 3/metabolismo
8.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31914380

RESUMEN

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Asunto(s)
Glucólisis , Interacciones Huésped-Patógeno , Interleucina-1beta/metabolismo , MicroARNs/metabolismo , Mycobacterium tuberculosis/fisiología , Fosfofructoquinasa-1/metabolismo , Animales , Antiinflamatorios/metabolismo , Secuencia de Bases , Proliferación Celular , Células HEK293 , Humanos , Interferón gamma/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , MicroARNs/genética , Fosfofructoquinasa-1/genética , Células RAW 264.7 , Tuberculosis/microbiología
9.
Vaccine ; 38(3): 635-643, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31727505

RESUMEN

Despite being in the midst of a global pandemic of infections caused by the pathogen Chlamydia trachomatis, a vaccine capable of inducing protective immunity remains elusive. Given the C. trachomatis mucosal port of entry, a formulation compatible with mucosal administration and capable of eliciting potent genital tract immunity is highly desirable. While subunit vaccines are considered safer and better tolerated, these are typically poorly immunogenic and require co-formulation with immune-potentiating adjuvants. However, of the adjuvants licensed for use in humans, very few drive robust cellular responses, a pre-requisite for protection against C. trachomatis infection. Recently, the cationic adjuvant formulations (CAF) have been shown to induce robust humoral and cellular immunity in pre-clinical models of chlamydia, malaria and tuberculosis (TB). Here, we demonstrate that CAF01 induces potent immune responses when combined with the major outer membrane protein (MOMP) of C. trachomatis following parenteral immunisation and also as part of a heterologous prime/boost regime. We show that a subcutaneous prime with CAF01-adjuvanted recombinant MOMP licenses antigen-specific immunity at distant mucosal sites which can be activated following oral antigen re-encounter in the absence of concomitant adjuvant stimulation. Finally, we shed light on the mechanism(s) through which CAF01 elicits robust antigen-specific immunity to co-formulated MOMP via type I interferon (IFN) signalling.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Chlamydia trachomatis/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Interferón Tipo I , Animales , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/prevención & control , Chlamydia trachomatis/inmunología , Composición de Medicamentos/métodos , Femenino , Inmunidad Celular/inmunología , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/inmunología
11.
Immunity ; 50(6): 1412-1424.e6, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31076360

RESUMEN

Assembly of inflammasomes after infection or injury leads to the release of interleukin-1ß (IL-1ß) and to pyroptosis. After inflammasome activation, cells either pyroptose or enter a hyperactivated state defined by IL-1ß secretion without cell death, but what controls these different outcomes is unknown. Here, we show that removal of the Toll-IL-1R protein SARM from macrophages uncouples inflammasome-dependent cytokine release and pyroptosis, whereby cells displayed increased IL-1ß production but reduced pyroptosis. Correspondingly, increasing SARM in cells caused less IL-1ß release and more pyroptosis. SARM suppressed IL-1ß by directly restraining the NLRP3 inflammasome and, hence, caspase-1 activation. Consistent with a role for SARM in pyroptosis, Sarm1-/- mice were protected from lipopolysaccharide (LPS)-stimulated sepsis. Pyroptosis-inducing, but not hyperactivating, NLRP3 stimulants caused SARM-dependent mitochondrial depolarization. Thus, SARM-dependent mitochondrial depolarization distinguishes NLRP3 activators that cause pyroptosis from those that do not, and SARM modulation represents a cell-intrinsic mechanism to regulate cell fate after inflammasome activation.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Citocinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas del Dominio Armadillo/genética , Biomarcadores , Supervivencia Celular , Proteínas del Citoesqueleto/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Unión Proteica , Piroptosis , Transducción de Señal
12.
J Immunol ; 202(4): 1145-1152, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30642984

RESUMEN

The cytokine IL-33 is a well-established inducer of Th2 responses. However, roles for IL-33 in promoting CD8, Th1, and T regulatory cell responses have also emerged. In this study, the role of IL-33 as a regulator of particulate vaccine adjuvant-induced Ag-specific cellular immunity was investigated. We found that polymeric nanoparticles surpassed alum in their ability to enhance Ag-specific CD8 and Th1 responses. IL-33 was a potent negative regulator of both CD8+ T cell and Th1 responses following i.m. vaccination with Ag and nanoparticles, whereas the cytokine was required for the nanoparticle enhancement in Ag-specific IL-10. In contrast to the effect on cellular immunity, Ab responses were comparable between vaccinated wild-type and IL-33-deficient mice. IL-33 did not compromise alum-induced adaptive cellular immunity after i.m. vaccination. These data suggest that IL-33 attenuates the induction of cellular immune responses by nanoparticulate adjuvants and should be considered in the rational design of vaccines targeting enhanced CD8 and Th1 responses.


Asunto(s)
Antígenos/inmunología , Inmunidad Celular/inmunología , Interleucina-33/inmunología , Vacunas/inmunología , Animales , Antígenos/administración & dosificación , Inyecciones Intramusculares , Interleucina-33/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/administración & dosificación , Nanopartículas/química , Vacunación , Vacunas/administración & dosificación
13.
FEBS J ; 285(13): 2377-2401, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29656546

RESUMEN

Growing awareness of the multiplicity of roles for the IL-1 family in immune regulation has prompted research exploring these cytokines in the context of vaccine-induced immunity. While tightly regulated, cytokines of the IL-1 family are normally released in response to cellular stress and in combination with other danger-/damage-associated molecular patterns (DAMPs), triggering potent local and systemic immune responses. In the context of infection or autoimmunity, engagement of IL-1 family receptors links robust innate responses to adaptive immunity. Clinical and experimental evidence has revealed that many vaccine adjuvants induce the release of one or multiple IL-1 family cytokines. The coordinated release of IL-1 family members in response to adjuvant-induced damage or cell death may be a determining factor in the transition from local inflammation to the induction of an adaptive response. Here, we analyse the effects of IL-1 family cytokines on innate and adaptive immunity with a particular emphasis on activation of antigen-presenting cells and induction of T cell-mediated immunity, and we address in detail the contribution of these cytokines to the modes of action of vaccine adjuvants including those currently approved for human use.


Asunto(s)
Inmunidad Adaptativa/inmunología , Citocinas/inmunología , Inmunidad Innata/inmunología , Interleucina-1/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Citocinas/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Interleucina-1/metabolismo , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
14.
Eur J Immunol ; 48(4): 705-715, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29349774

RESUMEN

The effectiveness of many vaccines licensed for clinical use relates to the induction of neutralising antibodies, facilitated by the inclusion of vaccine adjuvants, particularly alum. However, the ability of alum to preferentially promote humoral rather than cellular, particularly Th1-type responses, is not well understood. We demonstrate that alum activates immunosuppressive mechanisms following vaccination, which limit its capacity to induce Th1 responses. One of the key cytokines limiting excessive immune responses is IL-10. Injection of alum primed draining lymph node cells for enhanced IL-10 secretion ex vivo. Moreover, at the site of injection, macrophages and dendritic cells were key sources of IL-10 expression. Alum strongly enhanced the transcription and secretion of IL-10 by macrophages and dendritic cells. The absence of IL-10 signalling did not compromise alum-induced cell infiltration into the site of injection, but resulted in enhanced antigen-specific Th1 responses after vaccination. In contrast to its decisive regulatory role in regulating Th1 responses, there was no significant change in antigen-specific IgG1 antibody production following vaccination with alum in IL-10-deficient mice. Overall, these findings indicate that injection of alum promotes IL-10, which can block Th1 responses and may explain the poor efficacy of alum as an adjuvant for inducing protective Th1 immunity.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Compuestos de Alumbre/farmacología , Células Dendríticas/inmunología , Interleucina-10/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células TH1/inmunología , Animales , Células Cultivadas , Escherichia coli/inmunología , Femenino , Interleucina-10/biosíntesis , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunas/inmunología
15.
Nat Cell Biol ; 19(4): 267-269, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28361938

RESUMEN

The Hippo pathway responds to environmental factors including nutrient availability, cell density and substrate stiffness to regulate organ size. This pathway is now shown to also regulate antiviral defence by modulating the TBK1-mediated control of interferon production.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Transducción de Señal , Antivirales , Tamaño de los Órganos
16.
Infect Immun ; 85(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27872237

RESUMEN

Signaling through Toll-like receptors (TLRs), the main receptors in innate immunity, is essential for the defense of mucosal surfaces. It was previously shown that systemic TLR5 stimulation by bacterial flagellin induces an immediate, transient interleukin-22 (IL-22)-dependent antimicrobial response to bacterial or viral infections of the mucosa. This process was dependent on the activation of type 3 innate lymphoid cells (ILCs). The objective of the present study was to analyze the effects of flagellin treatment in a murine model of oral infection with Yersinia pseudotuberculosis (an invasive, Gram-negative, enteropathogenic bacterium that targets the small intestine). We found that systemic administration of flagellin significantly increased the survival rate after intestinal infection (but not systemic infection) by Y. pseudotuberculosis This protection was associated with a low bacterial count in the gut and the spleen. In contrast, no protection was afforded by administration of the TLR4 agonist lipopolysaccharide, suggesting the presence of a flagellin-specific effect. Lastly, we found that TLR5- and MyD88-mediated signaling was required for the protective effects of flagellin, whereas neither lymphoid cells nor IL-22 was involved.


Asunto(s)
Flagelina/inmunología , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Flagelina/administración & dosificación , Interleucinas/genética , Mucosa Intestinal/microbiología , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión , Transducción de Señal , Receptores Toll-Like/metabolismo , Infecciones por Yersinia pseudotuberculosis/microbiología , Infecciones por Yersinia pseudotuberculosis/mortalidad , Interleucina-22
17.
Future Microbiol ; 11: 1167-77, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27546231

RESUMEN

AIM: To evaluate efficacy of sublingual flagellin to treat acute pneumonia. MATERIALS & METHODS: Mice were treated sublingually with flagellin and challenged intranasally with a lethal dose of pneumococcus. Flagellins lacking TLR5 or NLRC4 activation domains were used to assess their contribution to protection. RESULTS: Sublingual flagellin protected mice in a TLR5-dependent, NLRC4-independent fashion. Neutrophils were required for protection. Flagellin-stimulated lung epithelial cells recapitulated the lung's transcriptional profile suggesting they could be targeted by flagellin in vivo. CONCLUSION: Ligation of TLR5, a pathogen recognition receptor not naturally engaged by pneumococcus, protects mice from invasive pneumonia when administered via sublingual route. This can be a highly cost-effective alternative therapy against pneumonia.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Proteínas de Unión al Calcio/inmunología , Flagelina/inmunología , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/prevención & control , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 5/inmunología , Administración Sublingual , Animales , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/química , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas de Unión al Calcio/genética , Femenino , Flagelina/administración & dosificación , Flagelina/química , Flagelina/genética , Humanos , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Neumonía Neumocócica/genética , Neumonía Neumocócica/microbiología , Dominios Proteicos , Streptococcus pneumoniae/genética , Receptor Toll-Like 5/genética
18.
Methods Mol Biol ; 1417: 1-43, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27221479

RESUMEN

For many years innate immunity was regarded as a relatively nonspecific set of mechanisms serving as a first line of defence to contain infections while the more refined adaptive immune response was developing. The discovery of pattern recognition receptors (PRRs) revolutionised the prevailing view of innate immunity, revealing its intimate connection with adaptive immunity and generation of effector and memory T- and B-cell responses. Among the PRRs, families of Toll-like receptors (TLRs), C-type lectin receptors (CLR), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) and nucleotide-binding domain, leucine-rich repeat-containing protein receptors (NLRs), along with a number of cytosolic DNA sensors and the family of absent in melanoma (AIM)-like receptors (ALRs), have been characterised. NLR sensors have been a particular focus of attention, and some NLRs have emerged as key orchestrators of the inflammatory response through the formation of large multiprotein complexes termed inflammasomes. However, several other functions not related to inflammasomes have also been described for NLRs. This chapter introduces the different families of PRRs, their signalling pathways, cross-regulation and their roles in immunosurveillance. The structure and function of NLRs is also discussed with particular focus on the non-inflammasome NLRs.


Asunto(s)
Inmunidad Innata , Receptores de Reconocimiento de Patrones/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Regulación de la Expresión Génica , Humanos , Inflamasomas/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología
19.
Immunity ; 44(3): 597-608, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944200

RESUMEN

The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Additionally, this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and immunoglobulin G2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Quitosano/administración & dosificación , Células Dendríticas/fisiología , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Nucleotidiltransferasas/metabolismo , Células TH1/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular , Células Cultivadas , ADN/metabolismo , Células Dendríticas/efectos de los fármacos , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunoglobulina G/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Nucleotidiltransferasas/genética , Especies Reactivas de Oxígeno/metabolismo , Vacunas/administración & dosificación
20.
Immunity ; 44(2): 368-79, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26885859

RESUMEN

Humans that are heterozygous for the common S180L polymorphism in the Toll-like receptor (TLR) adaptor Mal (encoded by TIRAP) are protected from a number of infectious diseases, including tuberculosis (TB), whereas those homozygous for the allele are at increased risk. The reason for this difference in susceptibility is not clear. We report that Mal has a TLR-independent role in interferon-gamma (IFN-γ) receptor signaling. Mal-dependent IFN-γ receptor (IFNGR) signaling led to mitogen-activated protein kinase (MAPK) p38 phosphorylation and autophagy. IFN-γ signaling via Mal was required for phagosome maturation and killing of intracellular Mycobacterium tuberculosis (Mtb). The S180L polymorphism, and its murine equivalent S200L, reduced the affinity of Mal for the IFNGR, thereby compromising IFNGR signaling in macrophages and impairing responses to TB. Our findings highlight a role for Mal outside the TLR system and imply that genetic variation in TIRAP may be linked to other IFN-γ-related diseases including autoimmunity and cancer.


Asunto(s)
Interferón gamma/metabolismo , Macrófagos/fisiología , Glicoproteínas de Membrana/metabolismo , Mycobacterium tuberculosis/inmunología , Receptores de Interleucina-1/metabolismo , Tuberculosis Pulmonar/inmunología , Animales , Autofagia/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Células HEK293 , Humanos , Inmunidad Innata/genética , Sistema de Señalización de MAP Quinasas/genética , Macrófagos/microbiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Polimorfismo Genético , Unión Proteica/genética , ARN Interferente Pequeño/genética , Receptores de Interferón/metabolismo , Receptores de Interleucina-1/genética , Tuberculosis Pulmonar/genética , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...