Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35074921

RESUMEN

Proinflammatory cytokine production by innate immune cells plays a crucial role in inflammatory diseases, but the molecular mechanisms controlling the inflammatory responses are poorly understood. Here, we show that TANK-binding kinase 1 (TBK1) serves as a vital regulator of proinflammatory macrophage function and protects against tissue inflammation. Myeloid cell-conditional Tbk1 knockout (MKO) mice spontaneously developed adipose hypertrophy and metabolic disorders at old ages, associated with increased adipose tissue M1 macrophage infiltration and proinflammatory cytokine expression. When fed with a high-fat diet, the Tbk1-MKO mice also displayed exacerbated hepatic inflammation and insulin resistance, developing symptoms of nonalcoholic steatohepatitis. Furthermore, myeloid cell-specific TBK1 ablation exacerbates inflammation in experimental colitis. Mechanistically, TBK1 functions in macrophages to suppress the NF-κB and MAP kinase signaling pathways and thus attenuate induction of proinflammatory cytokines, particularly IL-1ß. Ablation of IL-1 receptor 1 (IL-1R1) eliminates the inflammatory symptoms of Tbk1-MKO mice. These results establish TBK1 as a pivotal anti-inflammatory mediator that restricts inflammation in different disease models.


Asunto(s)
Inflamación/etiología , Inflamación/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Biomarcadores , Colitis/etiología , Colitis/metabolismo , Colitis/patología , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Regulación de la Expresión Génica , Glucosa/metabolismo , Hipertrofia , Inmunomodulación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Interleucina-1/deficiencia , Transducción de Señal
2.
Blood Adv ; 6(1): 200-206, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34555844

RESUMEN

Development of normal blood cells is often suppressed in juvenile myelomonocytic leukemia (JMML), a myeloproliferative neoplasm (MPN) of childhood, causing complications and impacting therapeutic outcomes. However, the mechanism underlying this phenomenon remains uncharacterized. To address this question, we induced the most common mutation identified in JMML (Ptpn11E76K) specifically in the myeloid lineage with hematopoietic stem cells (HSCs) spared. These mice uniformly developed a JMML-like MPN. Importantly, HSCs in the same bone marrow (BM) microenvironment were aberrantly activated and differentiated at the expense of self-renewal. As a result, HSCs lost quiescence and became exhausted. A similar result was observed in wild-type (WT) donor HSCs when co-transplanted with Ptpn11E76K/+ BM cells into WT mice. Co-culture testing demonstrated that JMML/MPN cells robustly accelerated differentiation in mouse and human normal hematopoietic stem/progenitor cells. Cytokine profiling revealed that Ptpn11E76K/+ MPN cells produced excessive IL-1ß, but not IL-6, T NF-α, IFN-γ, IL-1α, or other inflammatory cytokines. Depletion of the IL-1ß receptor effectively restored HSC quiescence, normalized their pool size, and rescued them from exhaustion in Ptpn11E76K/+/IL-1R-/- double mutant mice. These findings suggest IL-1ß signaling as a potential therapeutic target for preserving normal hematopoietic development in JMML.


Asunto(s)
Células Madre Hematopoyéticas , Inflamación , Interleucina-1beta , Leucemia Mielomonocítica Juvenil , Animales , Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Leucemia Mielomonocítica Juvenil/inmunología , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/patología , Ratones , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Receptores de Interleucina-1/deficiencia , Microambiente Tumoral
3.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941658

RESUMEN

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Asunto(s)
Infecciones por Bacteroides/inmunología , Bacteroides fragilis/inmunología , Fibras de la Dieta/efectos adversos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Peritonitis/inmunología , Animales , Infecciones por Bacteroides/microbiología , Dieta , Fibras de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Peritonitis/microbiología , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
4.
Clin Exp Dermatol ; 45(8): 967-973, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32882069

RESUMEN

The systemic autoinflammatory disorders (SAIDS) or periodic fever syndromes are disorders of innate immunity, which can be inherited or acquired. They are almost all very rare and easily overlooked; typically, patients will have seen multiple specialities prior to diagnosis, so a high level of clinical suspicion is key. It is important to note that these are 'high-value' diagnoses as the majority of these syndromes can be very effectively controlled, dramatically improving quality of life and providing protection against the development of irreversible complications such as AA amyloidosis. In Part 1 of this review, we took an overview of SAIDS and described the common features; in this article, we take a more in-depth look at the better recognized or more dermatologically relevant conditions.


Asunto(s)
Amiloidosis/prevención & control , Dermatólogos/estadística & datos numéricos , Enfermedades Autoinflamatorias Hereditarias/inmunología , Enfermedades del Sistema Inmune/inmunología , Receptores de Interleucina-1/deficiencia , Amiloidosis/etiología , Amiloidosis/patología , Antiinflamatorios no Esteroideos/uso terapéutico , Productos Biológicos/uso terapéutico , Colchicina/uso terapéutico , Síndromes Periódicos Asociados a Criopirina/diagnóstico , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Síndromes Periódicos Asociados a Criopirina/genética , Síndromes Periódicos Asociados a Criopirina/patología , Fiebre Mediterránea Familiar/diagnóstico , Fiebre Mediterránea Familiar/tratamiento farmacológico , Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/patología , Fiebre/diagnóstico , Fiebre/tratamiento farmacológico , Fiebre/genética , Fiebre/patología , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Enfermedades Autoinflamatorias Hereditarias/tratamiento farmacológico , Enfermedades Autoinflamatorias Hereditarias/genética , Enfermedades Autoinflamatorias Hereditarias/patología , Humanos , Enfermedades del Sistema Inmune/complicaciones , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/patología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/patología , Proteína Antagonista del Receptor de Interleucina 1/efectos adversos , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1/inmunología , Interleucina-1/metabolismo , Deficiencia de Mevalonato Quinasa/diagnóstico , Deficiencia de Mevalonato Quinasa/tratamiento farmacológico , Deficiencia de Mevalonato Quinasa/genética , Deficiencia de Mevalonato Quinasa/patología , Receptores de Interleucina-1/efectos de los fármacos , Receptores de Interleucina-1/genética , Receptores de Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/uso terapéutico , Síndrome de Schnitzler/diagnóstico , Síndrome de Schnitzler/tratamiento farmacológico , Síndrome de Schnitzler/inmunología , Síndrome de Schnitzler/patología , Enfermedades Cutáneas Genéticas/diagnóstico , Enfermedades Cutáneas Genéticas/inmunología , Enfermedades Cutáneas Genéticas/patología , Esteroides/uso terapéutico , Moduladores de Tubulina/uso terapéutico
5.
Cell Rep ; 32(3): 107911, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32698010

RESUMEN

Effector, but not naïve, T cells are activated by toll-like receptor-2 (TLR2) stimulation, leading to cytokine production and proliferation. We found that the differential response is attributable to the lack of expression of the adaptor protein TIRAP in naive T cells. TIRAP expression is induced upon T-cell receptor (TCR) stimulation and sustained by strong interleukin-2 (IL-2) signals. Expression of TIRAP requires TCR- and IL-2-induced mTORC1 activation. TLR2 stimulation induced the activation of nuclear factor κB (NF-κB) and ERK, leading to much higher production of interferon-γ (IFN-γ) by T helper 1 (Th1) cells cultured in a high concentration of IL-2 than by those cultured in a low concentration of IL-2. In contrast, TLR2 stimulation induces mTORC1 activation through TIRAP, which is essential for TLR2-mediated IFN-γ production. These data demonstrate that the mTORC1 signal confers the response to TLR2 signaling by inducing TIRAP expression and that the TIRAP-mTORC1 axis is critical for TLR2-mediated IFN-γ production by effector T cells.


Asunto(s)
Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Células Cultivadas , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Glicoproteínas de Membrana/deficiencia , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores de Interleucina-1/deficiencia , Células TH1/inmunología , Regulación hacia Arriba
6.
J Exp Med ; 217(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32573694

RESUMEN

Chronic pain is a major comorbidity of chronic inflammatory diseases. Here, we report that the cytokine IL-1ß, which is abundantly produced during multiple sclerosis (MS), arthritis (RA), and osteoarthritis (OA) both in humans and in animal models, drives pain associated with these diseases. We found that the type 1 IL-1 receptor (IL-1R1) is highly expressed in the mouse and human by a subpopulation of TRPV1+ dorsal root ganglion neurons specialized in detecting painful stimuli, termed nociceptors. Strikingly, deletion of the Il1r1 gene specifically in TRPV1+ nociceptors prevented the development of mechanical allodynia without affecting clinical signs and disease progression in mice with experimental autoimmune encephalomyelitis and K/BxN serum transfer-induced RA. Conditional restoration of IL-1R1 expression in nociceptors of IL-1R1-knockout mice induced pain behavior but did not affect joint damage in monosodium iodoacetate-induced OA. Collectively, these data reveal that neuronal IL-1R1 signaling mediates pain, uncovering the potential benefit of anti-IL-1 therapies for pain management in patients with chronic inflammatory diseases.


Asunto(s)
Inflamación/metabolismo , Inflamación/patología , Neuronas/metabolismo , Dolor/metabolismo , Dolor/patología , Receptores de Interleucina-1/metabolismo , Adulto , Anciano , Animales , Artritis Reumatoide/patología , Conducta Animal , Enfermedad Crónica , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Miembro Posterior/patología , Humanos , Hiperalgesia/complicaciones , Hiperalgesia/patología , Inflamación/complicaciones , Interleucina-1beta/metabolismo , Articulación de la Rodilla/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Mieloides/metabolismo , Neuronas/patología , Nociceptores/metabolismo , Osteoartritis , Dolor/complicaciones , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Células Receptoras Sensoriales/metabolismo , Raíces Nerviosas Espinales/metabolismo , Raíces Nerviosas Espinales/patología , Canales Catiónicos TRPV/metabolismo
7.
Life Sci Alliance ; 3(6)2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32345660

RESUMEN

IL-36R signaling plays an important role in the pathogenesis of psoriasis. We ought to assess the specific function of IL-36R in keratinocytes for the pathology of Aldara-induced psoriasis-like dermatitis. Il36r ΔK mice presenting deletion of IL-36R in keratinocytes were similarly resistant to Aldara-induced ear inflammation as Il36r -/- mice, but acanthosis was only prevented in Il36r -/- mice. FACS analysis revealed that IL-36R signaling in keratinocytes is mandatory for early neutrophil infiltration in Aldara-treated ears. RNASeq and qRT-PCR experiments demonstrated the crucial role of IL-36R signaling in keratinocytes for induction of IL-23, IL-17, and IL-22 at early time points. Taken together, our results demonstrate that IL-36R signaling in keratinocytes plays a major role in the induction of Aldara-induced psoriasis-like dermatitis by triggering early production of IL-23/IL-17/IL-22 cytokines and neutrophil infiltration.


Asunto(s)
Erupciones por Medicamentos/inmunología , Interleucina-23/biosíntesis , Queratinocitos/inmunología , Otitis Externa/inmunología , Psoriasis/inmunología , Receptores de Interleucina-1/deficiencia , Transducción de Señal/genética , Administración Cutánea , Animales , Erupciones por Medicamentos/etiología , Erupciones por Medicamentos/metabolismo , Femenino , Eliminación de Gen , Imiquimod/administración & dosificación , Imiquimod/efectos adversos , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Otitis Externa/inducido químicamente , Psoriasis/inducido químicamente , Receptores de Interleucina-1/genética , Transducción de Señal/inmunología , Interleucina-22
8.
Arterioscler Thromb Vasc Biol ; 40(6): 1533-1542, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32268786

RESUMEN

OBJECTIVE: Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS: These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.


Asunto(s)
Clopidogrel/farmacocinética , Clopidogrel/uso terapéutico , Resistencia a Medicamentos , Obesidad/complicaciones , Receptores de Interleucina-1/fisiología , Animales , Trombosis de las Arterias Carótidas/prevención & control , Clopidogrel/farmacología , Sistema Enzimático del Citocromo P-450/genética , Diabetes Mellitus , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrinolíticos , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/enzimología , Obesidad/etiología , Obesidad/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria , Profármacos/farmacocinética , Profármacos/uso terapéutico , Receptores de Interleucina-1/deficiencia
9.
PLoS One ; 15(3): e0230718, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210474

RESUMEN

Chlamydia trachomatis is the most common bacterial sexually-transmitted infection and the major cause of preventable blindness worldwide. The asymptomatic nature of many infections along with uncontrolled inflammation leads to irreversible damage in the upper genital tract and the tarsal conjunctivae, with the major complications of infertility and chronic pelvic pain, and blindness, respectively. Inflammation must, therefore, be tightly regulated to avoid an unrestrained immune response. The genetic factors that regulate inflammation through Toll-like receptor (TLR) signaling pathways during C. trachomatis infection have not been fully characterized. SIGIRR (also known as IL-1R8 or TIR8) can regulate inflammation in response to various pathogens and diseases. However, nothing is known about its role during C. trachomatis infection. Expression of the pro-inflammatory chemokine, IL-8, was measured in epithelial cells infected with C. trachomatis. The effect of SIGIRR was determined by depleting SIGIRR or over-expressing SIGIRR in the epithelial cells before infection. Our results indicate that, in the absence of SIGIRR, epithelial cells induce higher levels of the pro-inflammatory chemokine, IL-8, in response to C. trachomatis infection. In addition, SIGIRR associates with MyD88 in both infected and uninfected infected cells. Collectively, our data demonstrate that SIGIRR functions as a negative regulator of the immune response to C. trachomatis infection. This finding provides insights into the immuno-pathogenesis of C. trachomatis that can be used to treat and identify individuals at risk of uncontrolled inflammation during infection.


Asunto(s)
Chlamydia trachomatis/fisiología , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/metabolismo , Chlamydia trachomatis/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica/inmunología , Silenciador del Gen , Células HeLa , Humanos , Interleucina-8/genética , Factor 88 de Diferenciación Mieloide/metabolismo , ARN Mensajero/genética , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética
10.
J Innate Immun ; 12(5): 387-398, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31851971

RESUMEN

Innate immune response is a universal mechanism against invading pathogens. Toll-like receptors (TLRs), being part of a first line of defense, are responsible for detecting a variety of microorganisms. Among them TLR9, which is localized in endosomes, acts as a sensor for unmethylated CpG motifs present in bacteria, DNA viruses (e.g., HSV-1), or fungi. TLRs differ from one another by the use of accessory proteins. MyD88 adapter-like (Mal) adapter molecule is considered a positive regulator of TLR2- and TLR4-dependent pathways. It has been reported that this adapter may also negatively control signal transduction induced by TLR3 anchored in the endosome membrane. So far, the role of Mal adapter protein in the TLR9 signaling pathways has not been clarified. We show for the first time that Mal is engaged in TLR9-de-pendent expression of genes encoding IFNß and TNFα in HSV-1-infected or CpG-C-treated macrophages and requires a noncanonical NF-κB pathway. Moreover, using inhibitor of ERK1/2 we confirmed involvement of these kinases in TLR9-dependent induction of IFNß and TNFα. Our study points to a new role of Mal in TLR9 signaling through a hitherto unknown mechanism whereby lack of Mal specifically impairs ERK1/2-mediated induction of noncanonical NF-κB pathway and concomitant IFNß and TNFα production.


Asunto(s)
Herpesvirus Humano 1/fisiología , Interferón beta/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-1/metabolismo , Receptor Toll-Like 9/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Humanos , Macrófagos/virología , Glicoproteínas de Membrana/deficiencia , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Receptores de Interleucina-1/deficiencia , Transducción de Señal
11.
Cell Immunol ; 344: 103930, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31196568

RESUMEN

The tolerogenic dendritic cell dysfunction is associated with the pathogenesis of immune diseases. Microbial stimulus is required in the maintenance of immune functions. This study aims to elucidate the role of Mal signal in the maintenance of DEC205+ DC (decDC) immune tolerogenic function. In this study, peripheral DCs were collected from allergic rhinitis (AR) patients and healthy control (HC) subjects to assess the functional status of decDCs. An AR murine model was developed to test the role of Mal signals in the maintenance of decDCs' functions. We observed that AR decDCs (decDCs obtained from AR patients) were incompetent in the induction of type 1 regulatory T cells (Tr1 cells). AR decDCs expressed less IL-10 than that in HC decDCs. IL-10 mRNA decayed spontaneously in AR decDCs. Tat-activating regulatory DNA-binding protein-43 (TDP43) protected IL-10 mRNA from decay. AR decDCs expressed lower levels of Mal than that in HC decDCs. Mal depletion resulted in IL-10 mRNA decay in HC decDCs. Reconstitution of Mal in AR decDCs restored the capacity of inducing Tr1 cells and attenuated experimental AR in mice. In conclusion, Mal plays a critical role in the maintenance of decDC's immune tolerogenic function. The absence or insufficient Mal signal impairs decDC's tolerogenic property. Reconstitution of Mal in AR decDCs can restore the immune tolerogenic capacity, which may have translational potential in the treatment of AR and other allergic diseases.


Asunto(s)
Células Dendríticas/inmunología , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-1/metabolismo , Rinitis Alérgica/inmunología , Adulto , Animales , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Linfocitos T Reguladores/inmunología , Receptores Toll-Like/metabolismo
12.
Cancer Immunol Res ; 7(6): 874-885, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31018956

RESUMEN

Chronic inflammation, including that driven by autoimmunity, is associated with the development of B-cell lymphomas. IL1R8 is a regulatory receptor belonging to the IL1R family, which negatively regulates NF-κB activation following stimulation of IL1R or Toll-like receptor family members. IL1R8 deficiency is associated with the development of severe autoimmune lupus-like disease in lpr mice. We herein investigated whether concomitant exacerbated inflammation and autoimmunity caused by the deficiency of IL1R8 could recapitulate autoimmunity-associated lymphomagenesis. We thus monitored B-cell lymphoma development during the aging of IL1R8-deficient lpr mice, observing an increased lymphoid cell expansion that evolved to diffuse large B-cell lymphoma (DLBCL). Molecular and gene-expression analyses showed that the NF-κB pathway was constitutively activated in Il1r8 -/-/lpr B splenocytes. In human DLBCL, IL1R8 had reduced expression compared with normal B cells, and higher IL1R8 expression was associated with a better outcome. Thus, IL1R8 silencing is associated with increased lymphoproliferation and transformation in the pathogenesis of B-cell lymphomas associated with autoimmunity.


Asunto(s)
Autoinmunidad/genética , Susceptibilidad a Enfermedades , Linfoma/etiología , Receptores de Interleucina-1/deficiencia , Animales , Biomarcadores , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Inmunohistoquímica , Linfoma/metabolismo , Linfoma/patología , Linfoma de Células B Grandes Difuso/etiología , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
13.
J Neurotrauma ; 36(2): 370-379, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768967

RESUMEN

Important differences in the biology of focal and diffuse traumatic brain injury (TBI) subtypes may result in unique pathophysiological responses to shared molecular mechanisms. Interleukin-1 (IL-1) signaling has been tested as a potential therapeutic target in preclinical models of cerebral contusion and diffuse TBI, and in a phase II clinical trial, but no published studies have examined IL-1 signaling in an impact/acceleration closed head injury (CHI) model. We hypothesized that genetic deletion of IL-1 receptor-1 (IL-1R1 KO) would be beneficial in focal (contusion) and CHI in mice. Wild type and IL-1R1 KO mice were subjected to controlled cortical impact (CCI), or to CHI. CCI produced brain leukocyte infiltration, HMGB1 translocation and release, edema, cell death, and cognitive deficits. CHI induced peak rotational acceleration of 9.7 × 105 ± 8.1 × 104 rad/s2, delayed time to righting reflex, and robust Morris water maze deficits without deficits in tests of anxiety, locomotion, sensorimotor function, or depression. CHI produced no discernable acute plasmalemma damage or cell death, blood-brain barrier permeability to IgG, or brain edema and only a modest increase in brain leukocyte infiltration at 72 h. In both models, mature (17 kDa) interleukin-1 beta (IL-1ß) was induced by 24 h in CD31+ endothelial cells isolated from injured brain but was not induced in CD11b+ cells in either model. High mobility group box protein-1 was released from injured brain cells in CCI but not CHI. Surprisingly, cognitive outcome in mice with global deletion of IL-1R1 was improved in CHI, but worse after CCI without affecting lesion size, edema, or infiltration of CD11b+/CD45+ leukocytes in CCI. IL-1R1 may induce unique biological responses, beneficial or detrimental to cognitive outcome, after TBI depending on the pathoanatomical subtype. Brain endothelium is a hitherto unrecognized source of mature IL-1ß in both models.


Asunto(s)
Conmoción Encefálica/metabolismo , Conmoción Encefálica/patología , Contusión Encefálica/metabolismo , Contusión Encefálica/patología , Receptores de Interleucina-1/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-1/deficiencia
14.
J Interferon Cytokine Res ; 38(10): 445-451, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30328795

RESUMEN

Obesity is a prominent risk factor for colorectal cancer (CRC). One mechanism by which obesity promotes the development of CRC is by generating a chronic, low-grade state of colonic inflammation. Interleukin-1ß (IL-1ß), a proinflammatory cytokine often elevated in obesity, is known to activate several procarcinogenic signaling pathways that are implicated in colonic carcinogenesis. We therefore sought to define the role of IL-1ß in mediating some of the early biochemical and molecular events leading up to obesity-promoted CRC. Twenty-five wild-type (WT) C57BL/6J mice and 24 lacking a functional IL-1 receptor (IL1R-/-) were each randomized to either low-fat or high-fat diets, resulting in lean and obese mice. Compared to WT lean controls, WT obese mice displayed 30%-80% greater concentrations of IL-1ß and tumor necrosis factor-α (TNF-α) in the colonic mucosa (IL-1ß: P = 0.04; TNF-α: P < 0.05), activation of the Wnt signaling cascade [evidenced by a 2-fold increase in colonic crypt cells displaying intranuclear ß-catenin (P < 0.03)], and a significant expansion of the proliferation zone of the colonic crypt (P < 0.04). These obesity-induced alterations in colonic cytokines, Wnt signaling, and proliferation were absent in the obese IL1R-/- mice. In the absence of IL-1 signaling, obesity-induced elevations of colonic IL-1ß, TNF-α, Wnt activation, and enhanced epithelial proliferation no longer occur. These observations underscore the important mechanistic roles that IL-1 signaling appears to play in mediating the procancerous effects of obesity in the colon, thereby identifying a potential target for future strategies aimed at chemoprevention.


Asunto(s)
Colon/inmunología , Células Epiteliales/inmunología , Inflamación/inmunología , Interleucina-1/inmunología , Obesidad/inmunología , Transducción de Señal/inmunología , Animales , Proliferación Celular , Colon/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/inmunología , Vía de Señalización Wnt/inmunología
15.
Molecules ; 23(6)2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-29799517

RESUMEN

The ABP dendrimer, which is built on a phosphorus-based scaffold and bears twelve azabisphosphonate groups at its surface, is one of the dendrimers that has been shown to display immuno-modulatory and anti-inflammatory effects towards the human immune system. Its anti-inflammatory properties have been successfully challenged in animal models of inflammatory disorders. In this review, we trace the discovery and the evaluation of the therapeutic effects of the ABP dendrimer in three different animal models of both acute and chronic inflammatory diseases. We emphasize that its therapeutic effects rely on the enhancement of the production of Interleukin-10, the paradigm of anti-inflammatory cytokines, by different subsets of immune cells, such as monocytes/macrophages and CD4+ T lymphocytes.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Reumatoide/tratamiento farmacológico , Linfocitos T CD4-Positivos/efectos de los fármacos , Dendrímeros/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Interleucina-10/inmunología , Animales , Antiinflamatorios/síntesis química , Antígenos CD/genética , Antígenos CD/inmunología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Linfocitos T CD4-Positivos/inmunología , Dendrímeros/síntesis química , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Expresión Génica , Humanos , Interleucina-10/genética , Activación de Linfocitos/efectos de los fármacos , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Relación Estructura-Actividad
16.
J Interferon Cytokine Res ; 38(3): 137-144, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29565745

RESUMEN

This study aimed to evaluate the role of eplerenone on the modulation of interleukin (IL)-1ß and IL-33/sST2 signaling pathway in an experimental model of left ventricular (LV) systolic dysfunction after acute myocardial infarction (MI). MI rats were randomly assigned to no treatment (MI group, n = 10), to receive eplerenone (Epl group, n = 10), or anakinra (Ana group, n = 10). LV function was assessed by echocardiography. IL-1ß, IL-33/sST2, and cardiac fibrosis biomarkers were analyzed by quantitative real-time reverse transcription polymerase chain reaction (PCR). Rats with MI showed significant reduction of LV systolic function, but treatment with eplerenone or anakinra improved left ventricular end-diastolic volume (LVEDV) and LVEDV/mass values. In the infarcted myocardium, compared with sham animals, the MI group had higher level of IL-33, sST2, and IL-1ß, as well as higher concentrations of markers of fibrosis and inflammation. Treatment with anakinra downregulated sST2 but with no effects on IL-33. Eplerenone reduced levels of sST2 and IL-1ß significantly. Both anakinra and eplerenone treatments were associated with lower levels of fibrosis and inflammatory markers. IL-1ß could induce expression of sST2, accelerating the progression of heart failure after acute MI. Eplerenone could improve LV function by reducing expression of IL-1ß and sST2.


Asunto(s)
Antihipertensivos/farmacología , Eplerenona/farmacología , Interleucina-1beta/metabolismo , Interleucina-33/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Receptores de Interleucina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Disfunción Ventricular Izquierda/tratamiento farmacológico , Enfermedad Aguda , Animales , Interleucina-1beta/deficiencia , Masculino , Infarto del Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-1/deficiencia , Disfunción Ventricular Izquierda/metabolismo
17.
Kidney Int ; 93(3): 599-614, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29241623

RESUMEN

IL-36, a newly named member of the IL-1 cytokine family, includes 3 isoforms, IL-36α, IL-36ß, and IL-36γ, all of which bind to a heterodimer containing the IL-36 receptor (IL-36R). Little is known about the role of the IL-36 axis in acute kidney injury (AKI) pathogenesis. Therefore, we evaluated IL-36 function in the bilateral renal ischemia-reperfusion injury model of AKI using IL-36R knockout and wild-type mice. IL-36R was found to be expressed in the kidney, mainly in proximal tubules. In IL-36R knockout mice, plasma creatinine, blood urea nitrogen, and IL-6 levels after ischemia-reperfusion injury were significantly lower than those in wild-type mice. Immunohistological analysis revealed mild tubular injury. IL-36α/ß/γ levels were increased after ischemia-reperfusion injury, and IL-36α was expressed in lymphocytes and proximal tubular cells, but post-ischemia-reperfusion injury mRNA levels of IL-6 and TNF-α were low in IL-36R knockout mice. In primary cultures of renal tubular epithelial cells, IL-36α treatment upregulated NF-κB activity and Erk phosphorylation. Notably, in patients with AKI, urine IL-36α levels were increased, and IL-36α staining in renal biopsy samples was enhanced. Thus, IL-36α/IL-36R blockage could serve as a potential therapeutic target in AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Receptores de Interleucina-1/deficiencia , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Células Cultivadas , Citocinas/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Interleucina-1/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Fenotipo , Fosforilación , Receptores de Interleucina-1/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal
18.
Elife ; 62017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28347403

RESUMEN

Inflammation modifies risk and/or severity of a variety of brain diseases through still elusive molecular mechanisms. Here we show that hyperactivation of the interleukin 1 pathway, through either ablation of the interleukin 1 receptor 8 (IL-1R8, also known as SIGIRR or Tir8) or activation of IL-1R, leads to up-regulation of the mTOR pathway and increased levels of the epigenetic regulator MeCP2, bringing to disruption of dendritic spine morphology, synaptic plasticity and plasticity-related gene expression. Genetic correction of MeCP2 levels in IL-1R8 KO neurons rescues the synaptic defects. Pharmacological inhibition of IL-1R activation by Anakinra corrects transcriptional changes, restores MeCP2 levels and spine plasticity and ameliorates cognitive defects in IL-1R8 KO mice. By linking for the first time neuronal MeCP2, a key player in brain development, to immune activation and demonstrating that synaptic defects can be pharmacologically reversed, these data open the possibility for novel treatments of neurological diseases through the immune system modulation.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/fisiología , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Ratones , Ratones Noqueados , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética
19.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28235196

RESUMEN

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/deficiencia , Receptores de Interleucina-1/deficiencia , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Ácidos Teicoicos/metabolismo , Inmunidad Adaptativa , Niño , Femenino , Fibroblastos/metabolismo , Humanos , Inmunidad Innata , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/genética , Monocitos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Linaje , Fagocitos/metabolismo , Mutación Puntual , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Receptores de Interleucina-1/análisis , Receptores de Interleucina-1/genética , Infecciones Estafilocócicas/tratamiento farmacológico , Ácidos Teicoicos/inmunología , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
20.
Methods Mol Biol ; 1512: 171-188, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27885607

RESUMEN

Relevant animal models for Campylobacter jejuni infection have been difficult to establish due to C. jejuni's inability to cause disease in many common animal research models. Fortunately, recent work has proven successful in developing several new and relevant mouse models of C. jejuni infection, including the SIGIRR-deficient mouse strain that develops acute enterocolitis in response to C. jejuni. Here we describe how to properly infect mice with C. jejuni, as well as a number of accompanying histological techniques to aid in studying C. jejuni colonization and infection in mice.


Asunto(s)
Infecciones por Campylobacter/patología , Campylobacter jejuni/patogenicidad , Enterocolitis/patología , Inmunohistoquímica/métodos , Enfermedad Aguda , Factores de Edad , Animales , Infecciones por Campylobacter/genética , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/fisiología , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Enterocolitis/genética , Enterocolitis/microbiología , Femenino , Formaldehído/química , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Polímeros/química , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Factores Sexuales , Coloración y Etiquetado/métodos , Fijación del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...