Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nature ; 620(7976): 1063-1070, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587335

RESUMEN

High-grade serous ovarian cancers have low survival rates because of their late presentation with extensive peritoneal metastases and frequent chemoresistance1, and require new treatments guided by novel insights into pathogenesis. Here we describe the intrinsic tumour-suppressive activities of interferon-ε (IFNε). IFNε is constitutively expressed in epithelial cells of the fallopian tube, the cell of origin of high-grade serous ovarian cancers, and is then lost during development of these tumours. We characterize its anti-tumour activity in several preclinical models: ovarian cancer patient-derived xenografts, orthotopic and disseminated syngeneic models, and tumour cell lines with or without mutations in Trp53 and Brca genes. We use manipulation of the IFNε receptor IFNAR1 in different cell compartments, differential exposure status to IFNε and global measures of IFN signalling to show that the mechanism of the anti-tumour activity of IFNε involves direct action on tumour cells and, crucially, activation of anti-tumour immunity. IFNε activated anti-tumour T and natural killer cells and prevented the accumulation and activation of myeloid-derived suppressor cells and regulatory T cells. Thus, we demonstrate that IFNε is an intrinsic tumour suppressor in the female reproductive tract whose activities in models of established and advanced ovarian cancer, distinct from other type I IFNs, are compelling indications of potential new therapeutic approaches for ovarian cancer.


Asunto(s)
Interferón Tipo I , Neoplasias Ováricas , Proteínas Supresoras de Tumor , Animales , Femenino , Humanos , Línea Celular Tumoral , Células Epiteliales/metabolismo , Trompas Uterinas/metabolismo , Genes BRCA1 , Genes BRCA2 , Genes p53 , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Linfocitos T/inmunología , Linfocitos T Reguladores , Proteínas Supresoras de Tumor/inmunología , Proteínas Supresoras de Tumor/metabolismo
2.
Nat Immunol ; 16(4): 354-65, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25729923

RESUMEN

Interleukin 37 (IL-37) and IL-1R8 (SIGIRR or TIR8) are anti-inflammatory orphan members of the IL-1 ligand family and IL-1 receptor family, respectively. Here we demonstrate formation and function of the endogenous ligand-receptor complex IL-37-IL-1R8-IL-18Rα. The tripartite complex assembled rapidly on the surface of peripheral blood mononuclear cells upon stimulation with lipopolysaccharide. Silencing of IL-1R8 or IL-18Rα impaired the anti-inflammatory activity of IL-37. Whereas mice with transgenic expression of IL-37 (IL-37tg mice) with intact IL-1R8 were protected from endotoxemia, IL-1R8-deficient IL-37tg mice were not. Proteomic and transcriptomic investigations revealed that IL-37 used IL-1R8 to harness the anti-inflammatory properties of the signaling molecules Mer, PTEN, STAT3 and p62(dok) and to inhibit the kinases Fyn and TAK1 and the transcription factor NF-κB, as well as mitogen-activated protein kinases. Furthermore, IL-37-IL-1R8 exerted a pseudo-starvational effect on the metabolic checkpoint kinase mTOR. IL-37 thus bound to IL-18Rα and exploited IL-1R8 to activate a multifaceted intracellular anti-inflammatory program.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-18/inmunología , Interleucina-1/inmunología , Leucocitos Mononucleares/inmunología , Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1/genética , Subunidad alfa del Receptor de Interleucina-18/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-18/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Lipopolisacáridos/farmacología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/inmunología , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Tirosina Quinasa c-Mer
3.
Nat Immunol ; 14(9): 901-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23872679

RESUMEN

Type I interferons are important in regulating immune responses to pathogens and tumors. All interferons are considered to signal via the heterodimeric IFNAR1-IFNAR2 complex, yet some subtypes such as interferon-ß (IFN-ß) can exhibit distinct functional properties, although the molecular basis of this is unclear. Here we demonstrate IFN-ß can uniquely and specifically ligate to IFNAR1 in an IFNAR2-independent manner, and we provide the structural basis of the IFNAR1-IFN-ß interaction. The IFNAR1-IFN-ß complex transduced signals that modulated expression of a distinct set of genes independently of Jak-STAT pathways. Lipopolysaccharide-induced sepsis was ameliorated in Ifnar1(-/-) mice but not Ifnar2(-/-) mice, suggesting that IFNAR1-IFN-ß signaling is pathologically relevant. Thus, we provide a molecular basis for understanding specific functions of IFN-ß.


Asunto(s)
Interferón beta/química , Interferón beta/metabolismo , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Femenino , Lipopolisacáridos/efectos adversos , Ratones , Ratones Noqueados , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Receptor de Interferón alfa y beta/genética , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Choque Séptico/mortalidad
4.
J Biol Chem ; 293(9): 3168-3179, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29187603

RESUMEN

The type I interferons (IFNs) are a family of cytokines with diverse biological activities, including antiviral, antiproliferative, and immunoregulatory functions. The discovery of the hormonally regulated, constitutively expressed IFNϵ has suggested a function for IFNs in reproductive tract homeostasis and protection from infections, but its intrinsic activities are untested. We report here the expression, purification, and functional characterization of murine IFNϵ (mIFNϵ). Recombinant mIFNϵ (rmIFNϵ) exhibited an α-helical fold characteristic of type I IFNs and bound to IFNα/ß receptor 1 (IFNAR1) and IFNAR2, but, unusually, it had a preference for IFNAR1. Nevertheless, rmIFNϵ induced typical type I IFN signaling activity, including STAT1 phosphorylation and activation of canonical type I IFN signaling reporters, demonstrating that it uses the JAK-STAT signaling pathway. We also found that rmIFNϵ induces the activation of T, B, and NK cells and exhibits antiviral, antiproliferative, and antibacterial activities typical of type I IFNs, albeit with 100-1000-fold reduced potency compared with rmIFNα1 and rmIFNß. Surprisingly, although the type I IFNs generally do not display cross-species activities, rmIFNϵ exhibited high antiviral activity on human cells, suppressing HIV replication and inducing the expression of known HIV restriction factors in human lymphocytes. Our findings define the intrinsic properties of murine IFNϵ, indicating that it distinctly interacts with IFNAR and elicits pathogen-suppressing activity with a potency enabling host defense but with limited toxicity, appropriate for a protein expressed constitutively in a sensitive mucosal site, such as the reproductive tract.


Asunto(s)
Interferón Tipo I/química , Interferón Tipo I/metabolismo , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Proliferación Celular/efectos de los fármacos , Chlamydia/efectos de los fármacos , Femenino , Humanos , Inmunidad Mucosa , Interferón Tipo I/farmacología , Ratones , Fosforilación , Conformación Proteica en Hélice alfa , Células RAW 264.7 , Receptores de Interferón/metabolismo , Reproducción , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
5.
Nat Rev Immunol ; 7(3): 220-30, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17318233

RESUMEN

There is no immunological mechanism to adequately explain the sudden epidemic in allergies noted in the last 30 years in developed countries. The reduction in the development of allergic disorders observed in individuals infected with parasitic helminths, however, supports a possible role for worms in suppressing allergies. Helminths regulate the immunity of the host to ensure a mutually beneficial environment for the survival of both the parasite and the host. This interplay between helminths and allergic responses raises fundamental questions in immunobiology. Harnessing current mechanistic studies for translational research into helminth infections and atopy might have potential for the identification of novel biomarkers, and even therapeutics, in allergic diseases.


Asunto(s)
Helmintiasis/inmunología , Hipersensibilidad/inmunología , Hipersensibilidad/prevención & control , Tolerancia Inmunológica , Células Th2/inmunología , Células Th2/parasitología , Animales , Helmintiasis/parasitología , Humanos
6.
Hum Mol Genet ; 24(2): 436-49, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25209981

RESUMEN

Harlequin ichthyosis (HI) is a severe skin disease which leads to neonatal death in ∼50% of cases. It is the result of mutations in ABCA12, a protein that transports lipids required to establish the protective skin barrier needed after birth. To better understand the life-threatening newborn HI phenotype, we analysed the developing epidermis for consequences of lipid dysregulation in mouse models. We observed a pro-inflammatory signature which was characterized by chemokine upregulation in embryonic skin which is distinct from that seen in other types of ichthyosis. Inflammation also persisted in grafted HI skin. To examine the contribution of inflammation to disease development, we overexpressed interleukin-37b to globally suppress fetal inflammation, observing considerable improvements in keratinocyte differentiation. These studies highlight inflammation as an unexpected contributor to HI disease development in utero, and suggest that inhibiting inflammation may reduce disease severity.


Asunto(s)
Ictiosis Lamelar/embriología , Ictiosis Lamelar/inmunología , Animales , Diferenciación Celular , Quimiocinas/genética , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Epidermis/embriología , Epidermis/inmunología , Femenino , Humanos , Ictiosis Lamelar/genética , Ictiosis Lamelar/fisiopatología , Interleucina-1/genética , Interleucina-1/inmunología , Queratinocitos/citología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Piel/embriología , Piel/inmunología
7.
Immunol Cell Biol ; 95(5): 478-483, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28045025

RESUMEN

Interferon epsilon (IFNɛ) is a type I IFN that is expressed constitutively in the female reproductive tract (FRT), and contributes to protection in models of sexually transmitted infections. Using multiple cell systems, including reporter cell lines and activated peripheral blood lymphocytes (PBLs), we show that recombinant IFNɛ impairs HIV infection at stage(s) post HIV entry and up to the translation of viral proteins. Consistent with this, IFNɛ upregulated a number of host cell restriction factors that block HIV at these stages of the replication cycle. The potency of IFNɛ induction of these HIV restriction factors was comparable to conventional type I IFNs, namely IFNα and IFNß. IFNɛ also significantly reduced the infectivity of progeny virion particles likely by inducing expression of HIV restriction factors, such as IFITM3, which act at that stage of infection. Thus, our data demonstrate that human IFNɛ suppresses HIV replication at multiple stages of infection.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Interferones/metabolismo , Replicación Viral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/virología , Infecciones por VIH/patología , Células HeLa , Humanos , Interferón-alfa/metabolismo , Fitohemaglutininas/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Linfocitos T/virología , Virión/efectos de los fármacos , Virión/metabolismo , Replicación Viral/efectos de los fármacos
8.
J Immunol ; 192(9): 4425-35, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696235

RESUMEN

Circulating levels of a soluble type I IFNR are elevated in diseases, such as chronic inflammation, infections, and cancer, but whether it functions as an antagonist, agonist, or transporter is unknown. In this study, we elucidate the in vivo importance of the soluble type I IFNAR, soluble (s)IFNAR2a, which is generated by alternative splicing of the Ifnar2 gene. A transgenic mouse model was established to mimic the 10-15-fold elevated expression of sIFNAR2a observed in some human diseases. We generated transgenic mouse lines, designated SolOX, in which the transgene mRNA and protein-expression patterns mirrored the expression patterns of the endogenous gene. SolOX were demonstrated to be more susceptible to LPS-mediated septic shock, a disease model in which type I IFN plays a crucial role. This effect was independent of "classical" proinflammatory cytokines, such as TNF-α and IL-6, whose levels were unchanged. Because the increased levels of sIFNAR2a did not affect the kinetics of the increased interferonemia, this soluble receptor does not potentiate its ligand signaling by improving IFN pharmacokinetics. Mechanistically, increased levels of sIFNAR2a are likely to facilitate IFN signaling, as demonstrated in spleen cells overexpressing sIFNAR2a, which displayed quicker, higher, and more sustained activation of STAT1 and STAT3. Thus, the soluble IFNR is an important agonist of endogenous IFN actions in pathophysiological processes and also is likely to modulate the therapeutic efficacy of clinically administered IFNs.


Asunto(s)
Interferón Tipo I/inmunología , Receptor de Interferón alfa y beta/inmunología , Choque Séptico/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Immunoblotting , Inmunofenotipificación , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Interferón alfa y beta/metabolismo , Choque Séptico/metabolismo , Receptor Toll-Like 4/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(35): 14384-9, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23946428

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common lung disease of premature infants, with devastating short- and long-term consequences. The pathogenesis of BPD is multifactorial, but all triggers cause pulmonary inflammation. No therapy exists; therefore, we investigated whether the anti-inflammatory interleukin-1 receptor antagonist (IL-1Ra) prevents murine BPD. We precipitated BPD by perinatal inflammation (lipopolysaccharide injection to pregnant dams) and rearing pups in hyperoxia (65% or 85% O2). Pups were treated daily with IL-1Ra or vehicle for up to 28 d. Vehicle-injected animals in both levels of hyperoxia developed a severe BPD-like lung disease (alveolar number and gas exchange area decreased by up to 60%, alveolar size increased up to fourfold). IL-1Ra prevented this structural disintegration at 65%, but not 85% O2. Hyperoxia depleted pulmonary immune cells by 67%; however, extant macrophages and dendritic cells were hyperactivated, with CD11b and GR1 (Ly6G/C) highly expressed. IL-1Ra partially rescued the immune cell population in hyperoxia (doubling the viable cells), reduced the percentage that were activated by up to 63%, and abolished the unexpected persistence of IL-1α and IL-1ß on day 28 in hyperoxia/vehicle-treated lungs. On day 3, perinatal inflammation and hyperoxia each triggered a distinct pulmonary immune response, with some proinflammatory mediators increasing up to 20-fold and some amenable to partial or complete reversal with IL-1Ra. In summary, our analysis reveals a pivotal role for IL-1α/ß in murine BPD and an involvement for MIP (macrophage inflammatory protein)-1α and TREM (triggering receptor expressed on myeloid cells)-1. Because it effectively shields newborn mice from BPD, IL-1Ra emerges as a promising treatment for a currently irremediable disease that may potentially brighten the prognosis of the tiny preterm patients.


Asunto(s)
Displasia Broncopulmonar/prevención & control , Hiperoxia/complicaciones , Inflamación/complicaciones , Proteína Antagonista del Receptor de Interleucina 1/fisiología , Animales , Displasia Broncopulmonar/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Embarazo
10.
J Biol Chem ; 289(7): 4316-25, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24362029

RESUMEN

MicroRNA-155 (miR-155) is highly expressed in many cancers such as B cell lymphomas and myeloid leukemia and inflammatory disorders such as rheumatoid arthritis, atopic dermatitis, and multiple sclerosis. The role of miR-155 as both a promoter of inflammation and an oncogenic agent provides a clear need for miR-155 itself to be stringently regulated. We therefore investigated the transcriptional regulation of miR-155 in response to the respective pro- and anti-inflammatory mediators LPS and IL-10. Bioinformatic analysis revealed Ets binding sites on the miR-155 promoter, and we found that Ets2 is critical for miR-155 induction by LPS. Truncation and mutational analysis of the miR-155 promoter confirmed the role of the Ets2 binding site proximal to the transcription start site for LPS responsiveness. We observed increased binding of Ets2 to the miR-155 promoter and Ets2 deficient mice displayed decreased induction of miR-155 in response to LPS. IL-10 inhibited the induction of Ets2 mRNA and protein by LPS, thereby decreasing Ets2 function on the pri-155 promoter. We have thus identified Ets2 as a key novel regulator in both the positive and negative control of miR-155 in the inflammatory response.


Asunto(s)
Lipopolisacáridos/toxicidad , MicroARNs/biosíntesis , Proteína Proto-Oncogénica c-ets-2/metabolismo , Elementos de Respuesta , Animales , Línea Celular , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-10/biosíntesis , Interleucina-10/genética , Ratones , MicroARNs/genética , Proteína Proto-Oncogénica c-ets-2/genética
11.
Protein Expr Purif ; 94: 7-14, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24211771

RESUMEN

Interferon ß (IFNß) is a member of the type I interferon family of cytokines widely recognised for their anti-viral, anti-proliferative and immunomodulatory properties. Recombinant, biologically active forms of this cytokine are used clinically for the treatment of multiple sclerosis and in laboratories to study the role of this cytokine in health and disease. Established methods for expression of IFNß utilise either bacterial systems from which the insoluble recombinant proteins must be refolded, or mammalian expression systems in which large volumes of cell culture are required for recovery of acceptable yields. Utilising the baculovirus expression system and Trichoplusia ni (Cabbage Looper) BTI-TN-5B1-4 cell line, we report a reproducible method for production and purification of milligram/litre quantities of biologically active murine IFNß. Due to the design of our construct and the eukaryotic nature of insect cells, the resulting soluble protein is secreted allowing purification of the Histidine-tagged natively-folded protein from the culture supernatant. The IFNß purification method described is a two-step process employing immobilised metal-ion affinity chromatography (IMAC) and reverse-phase high performance liquid chromatography (RP-HPLC) that results in production of significantly more purified IFNß than any other reported eukaryotic-based expression system. Recombinant murine IFNß produced by this method was natively folded and demonstrated hallmark type I interferon biological effects including antiviral and anti-proliferative activities, and induced genes characteristic of IFNß activity in vivo. Recombinant IFNß also had specific activity levels exceeding that of the commercially available equivalent. Together, our findings provide a method for production of highly pure, biologically active murine IFNß.


Asunto(s)
Baculoviridae/genética , Regulación Viral de la Expresión Génica , Interferón beta/biosíntesis , Proteínas Recombinantes/biosíntesis , Animales , Baculoviridae/crecimiento & desarrollo , Línea Celular , Histidina/genética , Insectos/citología , Insectos/genética , Interferón beta/genética , Interferón beta/aislamiento & purificación , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación
12.
Cell Mol Gastroenterol Hepatol ; 17(2): 267-278, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37879406

RESUMEN

BACKGROUND & AIMS: Type I interferon (T1IFN) signalling is crucial for maintaining intestinal homeostasis. We previously found that the novel T1IFN, IFNε, is highly expressed by epithelial cells of the female reproductive tract, where it protects against pathogens. Its function has not been studied in the intestine. We hypothesize that IFNε is important in maintaining intestinal homeostasis. METHODS: We characterized IFNε expression in mouse and human intestine by immunostaining and studied its function in the dextran sulfate sodium (DSS) colitis model using both genetic knockouts and neutralizing antibody. RESULTS: We demonstrate that IFNε is expressed in human and mouse intestinal epithelium, and expression is lost in inflammation. Furthermore, we show that IFNε limits intestinal inflammation in mouse models. Regulatory T cell (Treg) frequencies were paradoxically decreased in DSS-treated IFNε-/- mice, suggesting a role for IFNε in maintaining the intestinal Treg compartment. Colitis was ameliorated by transfer of wild-type Tregs into IFNε-/- mice. This demonstrates that IFNε supports intestinal Treg function. CONCLUSIONS: Overall, we have shown IFNε expression in intestinal epithelium and its critical role in gut homeostasis. Given its known role in the female reproductive tract, we now show IFNε has a protective role across multiple mucosal surfaces.


Asunto(s)
Colitis , Humanos , Ratones , Femenino , Animales , Colitis/metabolismo , Mucosa Intestinal/metabolismo , Inflamación/metabolismo , Transducción de Señal , Interferones/metabolismo
13.
EMBO Mol Med ; 16(2): 267-293, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38263527

RESUMEN

The uterus is a unique mucosal site where immune responses are balanced to be permissive of a fetus, yet protective against infections. Regulation of natural killer (NK) cell responses in the uterus during infection is critical, yet no studies have identified uterine-specific factors that control NK cell responses in this immune-privileged site. We show that the constitutive expression of IFNε in the uterus plays a crucial role in promoting the accumulation, activation, and IFNγ production of NK cells in uterine tissue during Chlamydia infection. Uterine epithelial IFNε primes NK cell responses indirectly by increasing IL-15 production by local immune cells and directly by promoting the accumulation of a pre-pro-like NK cell progenitor population and activation of NK cells in the uterus. These findings demonstrate the unique features of this uterine-specific type I IFN and the mechanisms that underpin its major role in orchestrating innate immune cell protection against uterine infection.


Asunto(s)
Células Asesinas Naturales , Útero , Femenino , Humanos , Feto , Interferones
14.
J Immunol ; 186(8): 4631-9, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398616

RESUMEN

Parasitic-infection studies on rhesus macaque monkeys have shown juvenile animals to be more susceptible to infection than adults, but the immunological mechanism for this is not known. In this study, we investigated the age-dependent genesis of helminth-induced type 2 immune responses using adult (6-8-wk-old) and juvenile (21-28-d-old) mice. Following infection with the parasitic nematode Nippostrongylus brasiliensis, juvenile mice had increased susceptibility to infection relative to adult mice. Juvenile mice developed a delayed type 2 immune response with decreased Th2 cytokine production, IgE Ab responses, mouse mast cell protease 1 levels, and intestinal goblet cell induction. This innate immune defect in juvenile mice was independent of TLR signaling, dendritic cells, or CD4(+) cell function. Using IL-4-eGFP mice, it was demonstrated that the numbers of IL-4-producing basophil and eosinophils were comparable in young and adult naive mice; however, following helminth infection, the early induction of these cells was impaired in juvenile mice relative to older animals. In nonhelminth models, there was an innate in vivo defect in activation of basophils, but not eosinophils, in juvenile mice compared with adult animals. The specific role for basophils in this innate defect in helminth-induced type 2 immunity was confirmed by the capacity of adoptively transferred adult-derived basophils, but not eosinophils, to restore the ability of juvenile mice to expel N. brasiliensis. The defect in juvenile mice with regard to helminth-induced innate basophil-mediated type 2 response is relevant to allergic conditions.


Asunto(s)
Basófilos/inmunología , Inmunidad Innata/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Factores de Edad , Animales , Basófilos/metabolismo , Susceptibilidad a Enfermedades/inmunología , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interacciones Huésped-Parásitos/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-4/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/fisiología , Infecciones por Strongylida/parasitología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
15.
J Exp Med ; 203(4): 1105-16, 2006 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-16606668

RESUMEN

Type 2 immunity, which involves coordinated regulation of innate and adaptive immune responses, can protect against helminth parasite infection, but may lead to allergy and asthma after inappropriate activation. We demonstrate that il25(-/-) mice display inefficient Nippostrongylus brasiliensis expulsion and delayed cytokine production by T helper 2 cells. We further establish a key role for interleukin (IL)-25 in regulating a novel population of IL-4-, IL-5-, IL-13-producing non-B/non-T (NBNT), c-kit+, FcepsilonR1- cells during helminth infection. A deficit in this population in il25(-/-) mice correlates with inefficient N. brasiliensis expulsion. In contrast, administration of recombinant IL-25 in vivo induces the appearance of NBNT, c-kit+, FcepsilonR1- cells and leads to rapid worm expulsion that is T and B cell independent, but type 2 cytokine dependent. We demonstrate that these IL-25-regulated cells appear rapidly in the draining lymph nodes, implicating them as a source of type 2 cytokines during initiation of worm expulsion.


Asunto(s)
Linfocitos B/citología , Basófilos/metabolismo , Interleucinas/fisiología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Linfocitos T/citología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Citocinas/biosíntesis , Citocinas/clasificación , Interleucina-13/biosíntesis , Interleucina-13/deficiencia , Interleucina-13/genética , Interleucina-4/biosíntesis , Interleucina-4/deficiencia , Interleucina-4/genética , Interleucina-5/biosíntesis , Interleucina-5/deficiencia , Interleucina-5/genética , Interleucinas/administración & dosificación , Interleucinas/deficiencia , Interleucinas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de IgE/deficiencia , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Immunol Cell Biol ; 90(5): 510-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22430250

RESUMEN

The mucosal system is the first line of defense against many pathogens. It is continuously exposed to dietary and microbial antigens, and thus the host must maintain a homeostatic environment between commensal microbiota and pathogenic infections. Following infections and inflammatory events, a rapid innate immune response is evoked to dampen the inflammatory processes. Type I interferons, a family of pleiotropic cytokines and major products of the innate immune response, have a key role in these early immune events at the mucosa, as reviewed here. With the emergence of new discoveries of immune cell types in mucosal tissues and their reactions to commensal and pathogenic organisms, we also review the opportunities for exciting research in this field.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Mucosa , Interferón Tipo I/inmunología , Animales , Homeostasis/inmunología , Humanos , Inmunomodulación , Metagenoma/inmunología , Membrana Mucosa/inmunología
17.
JCI Insight ; 7(18)2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-35862222

RESUMEN

Although published studies have demonstrated that IFN-ε has a crucial role in regulating protective immunity in the mouse female reproductive tract, expression and regulation of IFN-ε in the human female reproductive tract (hFRT) have not been characterized to our knowledge. We obtained hFRT samples from a well-characterized cohort of women to enable us to comprehensively assess ex vivo IFN-ε expression in the hFRT at various stages of the menstrual cycle. We found that among the various types of IFNs, IFN-ε was uniquely, selectively, and constitutively expressed in the hFRT epithelium. It had distinct expression patterns in the surface and glandular epithelia of the upper hFRT compared with basal layers of the stratified squamous epithelia of the lower hFRT. There was cyclical variation of IFN-ε expression in the endometrial epithelium of the upper hFRT and not in the distal FRT, consistent with selective endometrial expression of the progesterone receptor and regulation of the IFNE promoter by progesterone. Because we showed IFN-ε stimulated important protective IFN-regulated genes in FRT epithelium, this characterization is a key element in understanding the mechanisms of hormonal control of mucosal immunity.


Asunto(s)
Endometrio , Inmunidad Innata , Interferones , Animales , Endometrio/inmunología , Epitelio/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Interferones/genética , Interferones/metabolismo , Ratones , Progesterona/metabolismo , Regiones Promotoras Genéticas , Receptores de Progesterona/metabolismo
18.
Sci Transl Med ; 14(639): eaaz8454, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35385341

RESUMEN

Postnatal maturation of the immune system is poorly understood, as is its impact on illnesses afflicting term or preterm infants, such as bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension. These are both cardiopulmonary inflammatory diseases that cause substantial mortality and morbidity with high treatment costs. Here, we characterized blood samples collected from 51 preterm infants longitudinally at five time points, 20 healthy term infants at birth and age 3 to 16 weeks, and 5 healthy adults. We observed strong associations between type 2 immune polarization in circulating CD3+CD4+ T cells and cardiopulmonary illness, with odds ratios up to 24. Maternal magnesium sulfate therapy, delayed hepatitis B vaccination, and increasing fetal, but not maternal, chorioamnionitis severity were associated with attenuated type 2 polarization. Blocking type 2 mediators such as interleukin-4 (IL-4), IL-5, IL-13, or signal transducer and activator of transcription 6 (STAT6) in murine neonatal cardiopulmonary disease in vivo prevented changes in cell type composition, increases in IL-1ß and IL-13, and losses of pulmonary capillaries, but not gains in larger vessels. Thereby, type 2 blockade ameliorated lung inflammation, protected alveolar and vascular integrity, and confirmed the pathological impact of type 2 cytokines and STAT6. In-depth flow cytometry and single-cell transcriptomics of mouse lungs further revealed complex associations between immune polarization and cardiopulmonary disease. Thus, this work advances knowledge on developmental immunology and its impact on early life disease and identifies multiple therapeutic approaches that may relieve inflammation-driven suffering in the youngest patients.


Asunto(s)
Displasia Broncopulmonar , Interleucina-13 , Animales , Displasia Broncopulmonar/etiología , Displasia Broncopulmonar/patología , Displasia Broncopulmonar/prevención & control , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Inflamación/complicaciones , Pulmón/patología , Ratones , Embarazo
19.
J Exp Med ; 202(10): 1319-25, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16301741

RESUMEN

The coevolution of humans and infectious agents has exerted selective pressure on the immune system to control potentially lethal infections. Correspondingly, pathogens have evolved with various strategies to modulate and circumvent the host's innate and adaptive immune response. Schistosoma species are helminth parasites with genes that have been selected to modulate the host to tolerate chronic worm infections, often for decades, without overt morbidity. The modulation of immunity by schistosomes has been shown to prevent a range of immune-mediated diseases, including allergies and autoimmunity. Individual immune-modulating schistosome molecules have, therefore, therapeutic potential as selective manipulators of the immune system to prevent unrelated diseases. Here we show that S. mansoni eggs secrete a protein into host tissues that binds certain chemokines and inhibits their interaction with host chemokine receptors and their biological activity. The purified recombinant S. mansoni chemokine binding protein (smCKBP) suppressed inflammation in several disease models. smCKBP is unrelated to host proteins and is the first described chemokine binding protein encoded by a pathogenic human parasite and may have potential as an antiinflammatory agent.


Asunto(s)
Quimiocinas/antagonistas & inhibidores , Quimiocinas/metabolismo , Proteínas del Helminto/metabolismo , Mediadores de Inflamación/metabolismo , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Proteínas del Helminto/genética , Humanos , Intestinos/parasitología , Intestinos/patología , Hígado/parasitología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Óvulo/metabolismo , Proteómica , Schistosoma mansoni/genética
20.
J Allergy Clin Immunol ; 125(5): 1114-1124.e8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20304473

RESUMEN

BACKGROUND: Parasitic helminth infections of humans have been shown to suppress the immune response to allergens. Experimentally, infection of mice with the helminth Schistosoma mansoni prevents allergic airway inflammation and anaphylaxis via IL-10 and B cells. OBJECTIVE: To identify and characterize the specific helminth-induced regulatory B-cell subpopulation and determine the mechanism by which these regulatory B cells suppress allergic airway inflammation. METHODS: IL-10-producing B cells from the spleens of helminth-infected mice were phenotyped, isolated, and transferred to ovalbumin-sensitized mice, and their ability to modulate allergic airway inflammation was analyzed. RESULTS: S mansoni infection induced IL-10-producing CD1d(high) regulatory B cells that could prevent ovalbumin-induced allergic airway inflammation following passive transfer to ovalbumin-sensitized recipients. The capacity of regulatory B cells to suppress allergic airway inflammation was dependent on the expression of CD1d, and they functioned via an IL-10-mediated mechanism. Regulatory B cells induced pulmonary infiltration of CD4(+)CD25(+) forkhead box protein 3(+) regulatory T cells, independent of TGF-beta, thereby suppressing allergic airway inflammation. Regulatory B cells that were generated ex vivo also suppressed the development of allergic airway inflammation. Furthermore, the transfer of regulatory B cells reversed established airway inflammation in ovalbumin-sensitized mice. CONCLUSION: We have generated in vivo and ex vivo a regulatory B cell that can prevent or reverse allergen-induced airway inflammation via regulatory T cells.


Asunto(s)
Asma/prevención & control , Linfocitos B/inmunología , Factores de Transcripción Forkhead/metabolismo , Hipersensibilidad/prevención & control , Inflamación/prevención & control , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD1d/metabolismo , Asma/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Hipersensibilidad/inmunología , Inflamación/inmunología , Interleucina-10/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Linfocitos T Reguladores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA