Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
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
2.
Immunity ; 45(6): 1285-1298, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27939673

RESUMEN

Allergic disease originates in early life and polymorphisms in interleukin-33 gene (IL33) and IL1RL1, coding for IL-33R and decoy receptor sST2, confer allergy risk. Early life T helper 2 (Th2) cell skewing and allergy susceptibility are often seen as remnants of feto-maternal symbiosis. Here we report that shortly after birth, innate lymphoid type 2 cells (ILC2s), eosinophils, basophils, and mast cells spontaneously accumulated in developing lungs in an IL-33-dependent manner. During the phase of postnatal lung alveolarization, house dust mite exposure further increased IL-33, which boosted cytokine production in ILC2s and activated CD11b+ dendritic cells (DCs). IL-33 suppressed IL-12p35 and induced OX40L in neonatal DCs, thus promoting Th2 cell skewing. Decoy sST2 had a strong preventive effect on asthma in the neonatal period, less so in adulthood. Thus, enhanced neonatal Th2 cell skewing to inhaled allergens results from postnatal hyperactivity of the IL-33 axis during a period of maximal lung remodeling.


Asunto(s)
Asma/inmunología , Interleucina-33/inmunología , Pulmón/crecimiento & desarrollo , Pulmón/inmunología , Células Th2/inmunología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipersensibilidad/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pyroglyphidae/inmunología , Transducción de Señal/inmunología
3.
J Immunol ; 211(4): 626-632, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37387671

RESUMEN

The transcription factor retinoic acid-related orphan receptor α (RORα) is important in regulating several physiological functions, such as cellular development, circadian rhythm, metabolism, and immunity. In two in vivo animal models of type 2 lung inflammation, Nippostrongylus brasiliensis infection and house dust mite (HDM) sensitization, we show a role for Rora in Th2 cellular development during pulmonary inflammation. N. brasiliensis infection and HDM challenge induced an increase in frequency of Rora-expressing GATA3+CD4 T cells in the lung. Using staggerer mice, which have a ubiquitous deletion of functional RORα, we generated bone marrow chimera mice, and we observed a delayed worm expulsion and reduced frequency in the expansion of Th2 cells and innate lymphoid type 2 cells (ILC2s) in the lungs after N. brasiliensis infection. ILC2-deficient mouse (Rorafl/flIl7raCre) also had delayed worm expulsion with associated reduced frequency of Th2 cells and ILC2s in the lungs after N. brasiliensis infection. To further define the role for Rora-expressing Th2 cells, we used a CD4-specific Rora-deficient mouse (Rorafl/flCD4Cre), with significantly reduced frequency of lung Th2 cells, but not ILC2, after N. brasiliensis infection and HDM challenge. Interestingly, despite the reduction in pulmonary Th2 cells in Rorafl/flCD4Cre mice, this did not impact the expulsion of N. brasiliensis after primary and secondary infection, or the generation of lung inflammation after HDM challenge. This study demonstrates a role for RORα in Th2 cellular development during pulmonary inflammation that could be relevant to the range of inflammatory diseases in which RORα is implicated.


Asunto(s)
Inmunidad Innata , Neumonía , Ratones , Animales , Células Th2 , Receptor alfa de Ácido Retinoico , Linfocitos T CD4-Positivos , Tretinoina
4.
Nat Immunol ; 13(3): 229-36, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22267218

RESUMEN

Nuocytes are essential in innate type 2 immunity and contribute to the exacerbation of asthma responses. Here we found that nuocytes arose in the bone marrow and differentiated from common lymphoid progenitors, which indicates they are distinct, previously unknown members of the lymphoid lineage. Nuocytes required interleukin 7 (IL-7), IL-33 and Notch signaling for development in vitro. Pro-T cell progenitors at double-negative stage 1 (DN1) and DN2 maintained nuocyte potential in vitro, although the thymus was not essential for nuocyte development. Notably, the transcription factor RORα was critical for the development of nuocytes and their role in the expulsion of parasitic worms.


Asunto(s)
Diferenciación Celular , Leucocitos/inmunología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Animales , Interleucina-7/inmunología , Interleucina-7/metabolismo , Leucocitos/citología , Leucocitos/metabolismo , Ratones , Nippostrongylus/inmunología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Transducción de Señal , Infecciones por Strongylida/inmunología , Timocitos/inmunología
5.
Nature ; 556(7699): 113-117, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29590092

RESUMEN

The endogenous metabolite itaconate has recently emerged as a regulator of macrophage function, but its precise mechanism of action remains poorly understood. Here we show that itaconate is required for the activation of the anti-inflammatory transcription factor Nrf2 (also known as NFE2L2) by lipopolysaccharide in mouse and human macrophages. We find that itaconate directly modifies proteins via alkylation of cysteine residues. Itaconate alkylates cysteine residues 151, 257, 288, 273 and 297 on the protein KEAP1, enabling Nrf2 to increase the expression of downstream genes with anti-oxidant and anti-inflammatory capacities. The activation of Nrf2 is required for the anti-inflammatory action of itaconate. We describe the use of a new cell-permeable itaconate derivative, 4-octyl itaconate, which is protective against lipopolysaccharide-induced lethality in vivo and decreases cytokine production. We show that type I interferons boost the expression of Irg1 (also known as Acod1) and itaconate production. Furthermore, we find that itaconate production limits the type I interferon response, indicating a negative feedback loop that involves interferons and itaconate. Our findings demonstrate that itaconate is a crucial anti-inflammatory metabolite that acts via Nrf2 to limit inflammation and modulate type I interferons.


Asunto(s)
Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Proteína 1 Asociada A ECH Tipo Kelch/química , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/metabolismo , Succinatos/metabolismo , Alquilación , Animales , Carboxiliasas , Bovinos , Cisteína/química , Cisteína/metabolismo , Citocinas/biosíntesis , Citocinas/inmunología , Retroalimentación Fisiológica , Femenino , Células HEK293 , Humanos , Hidroliasas/biosíntesis , Interferón beta/inmunología , Interferón beta/farmacología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Proteínas/metabolismo , Ratas , Ratas Wistar , Succinatos/química
6.
Immunity ; 41(2): 283-95, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25088770

RESUMEN

Group 2 innate lymphoid cells (ILC2s) release interleukin-13 (IL-13) during protective immunity to helminth infection and detrimentally during allergy and asthma. Using two mouse models to deplete ILC2s in vivo, we demonstrate that T helper 2 (Th2) cell responses are impaired in the absence of ILC2s. We show that MHCII-expressing ILC2s interact with antigen-specific T cells to instigate a dialog in which IL-2 production from T cells promotes ILC2 proliferation and IL-13 production. Deletion of MHCII renders IL-13-expressing ILC2s incapable of efficiently inducing Nippostrongylus brasiliensis expulsion. Thus, during transition to adaptive T cell-mediated immunity, the ILC2 and T cell crosstalk contributes to their mutual maintenance, expansion and cytokine production. This interaction appears to augment dendritic-cell-induced T cell activation and identifies a previously unappreciated pathway in the regulation of type-2 immunity.


Asunto(s)
Comunicación Celular/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Nippostrongylus/inmunología , Células Th2/inmunología , Animales , Presentación de Antígeno/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Inmunidad Celular , Inmunidad Innata , Interleucina-13/biosíntesis , Interleucina-13/metabolismo , Interleucina-2/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Immunity ; 40(1): 40-50, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24412616

RESUMEN

Fibrosis in response to tissue damage or persistent inflammation is a pathological hallmark of many chronic degenerative diseases. By using a model of acute peritoneal inflammation, we have examined how repeated inflammatory activation promotes fibrotic tissue injury. In this context, fibrosis was strictly dependent on interleukin-6 (IL-6). Repeat inflammation induced IL-6-mediated T helper 1 (Th1) cell effector commitment and the emergence of STAT1 (signal transducer and activator of transcription-1) activity within the peritoneal membrane. Fibrosis was not observed in mice lacking interferon-γ (IFN-γ), STAT1, or RAG-1. Here, IFN-γ and STAT1 signaling disrupted the turnover of extracellular matrix by metalloproteases. Whereas IL-6-deficient mice resisted fibrosis, transfer of polarized Th1 cells or inhibition of MMP activity reversed this outcome. Thus, IL-6 causes compromised tissue repair by shifting acute inflammation into a more chronic profibrotic state through induction of Th1 cell responses as a consequence of recurrent inflammation.


Asunto(s)
Interleucina-6/metabolismo , Peritoneo/patología , Peritonitis/genética , Peritonitis/patología , Células TH1/inmunología , Enfermedad Aguda , Traslado Adoptivo , Animales , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Retroalimentación Fisiológica , Fibrosis , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células TH1/trasplante
8.
Immunity ; 38(1): 66-78, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23177319

RESUMEN

Suppressors of cytokine signaling (SOCS) are important regulators of lipopolysaccharide (LPS) and cytokine responses but their role in macrophage polarization is unknown. We have shown here that myeloid-restricted Socs3 deletion (Socs3(Lyz2cre)) resulted in resistance to LPS-induced endotoxic shock, whereas Socs2(-/-) mice were highly susceptible. We observed striking bias toward M2-like macrophages in Socs3(Lyz2cre) mice, whereas the M1-like population was enriched in Socs2(-/-) mice. Adoptive transfer experiments showed that responses to endotoxic shock and polymicrobial sepsis were transferable and macrophage dependent. Critically, this dichotomous response was associated with enhanced regulatory T (Treg) cell recruitment by Socs3(Lyz2cre) cells, whereas Treg cell recruitment was absent in the presence of Socs2(-/-) macrophages. In addition, altered polarization coincided with enhanced interferon-gamma (IFN-γ)-induced signal transducer and activator of transcription-1 (STAT1) activation in Socs2(-/-) macrophages and enhanced interleukin-4 (IL-4) plus IL-13-induced STAT6 phosphorylation in Socs3(Lyz2cre) macrophages. SOCS, therefore, are essential controllers of macrophage polarization, regulating inflammatory responses.


Asunto(s)
Polaridad Celular/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Traslado Adoptivo , Animales , Regulación de la Expresión Génica , Interleucina-10/inmunología , Interleucina-10/metabolismo , Macrófagos/trasplante , Ratones , Factores de Transcripción STAT/metabolismo , Sepsis/genética , Sepsis/inmunología , Sepsis/prevención & control , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Isogénico
9.
Mol Cancer ; 19(1): 16, 2020 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-31987050

RESUMEN

Since the publication of this work [1] and in response to a recent query that was brought to our attention in relation to the Western Blot in Figure 1(C) for NP2, protein lysates prepared around the same time as those presented in the manuscript in question, were run by SDS-PAGE under similar experimental conditions and probed using the same primary antibodies to NP1 and NP2 that were used originally.

10.
Mol Biol Rep ; 47(2): 1491-1498, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31811500

RESUMEN

Safe harbor loci allow predicable integration of a transgene into the genome without perturbing endogenous gene activity and for decades have been exploited in the mouse to investigate gene function, generate humanised models and create tissue specific reporter and Cre recombinase expressing lines. Herein, we show that the murine Hipp11 intergenic region can facilitate highly efficient integration of a large transgene-the human CD1A promoter and coding region-by means of CRISPR-Cas9 mediated homology directed repair. The data shows that the single copy human CD1A transgene is faithfully expressed in an inducible manner in homozygous animals in both macrophage and dendritic cells. Our results validate the Hipp11 intergenic region as being a highly amenable target site for functional transgene integration in mouse.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , ADN Intergénico/genética , Expresión Génica , Transgenes , Animales , Antígenos CD1/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Sitios Genéticos , Genoma , Humanos , Ratones Transgénicos
11.
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
12.
J Immunol ; 199(2): 707-717, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28615416

RESUMEN

Atopic dermatitis (AD) is a common inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide and is associated with dysregulation of the skin barrier. Although type 2 responses are implicated in AD, emerging evidence indicates a potential role for the IL-17A signaling axis in AD pathogenesis. In this study we show that in the filaggrin mutant mouse model of spontaneous AD, IL-17RA deficiency (Il17ra-/- ) resulted in severe exacerbation of skin inflammation. Interestingly, Il17ra-/- mice without the filaggrin mutation also developed spontaneous progressive skin inflammation with eosinophilia, as well as increased levels of thymic stromal lymphopoietin (TSLP) and IL-5 in the skin. Il17ra-/- mice have a defective skin barrier with altered filaggrin expression. The barrier dysregulation and spontaneous skin inflammation in Il17ra-/- mice was dependent on TSLP, but not the other alarmins IL-25 and IL-33. The associated skin inflammation was mediated by IL-5-expressing pathogenic effector Th2 cells and was independent of TCRγδ T cells and IL-22. An absence of IL-17RA in nonhematopoietic cells, but not in the hematopoietic cells, was required for the development of spontaneous skin inflammation. Skin microbiome dysbiosis developed in the absence of IL-17RA, with antibiotic intervention resulting in significant amelioration of skin inflammation and reductions in skin-infiltrating pathogenic effector Th2 cells and TSLP. This study describes a previously unappreciated protective role for IL-17RA signaling in regulation of the skin barrier and maintenance of skin immune homeostasis.


Asunto(s)
Dermatitis Atópica/inmunología , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-17/metabolismo , Piel/crecimiento & desarrollo , Piel/patología , Animales , Citocinas/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Disbiosis , Eosinofilia/inmunología , Proteínas Filagrina , Regulación de la Expresión Génica , Homeostasis , Interleucina-33/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Interleucinas/genética , Interleucinas/inmunología , Proteínas de Filamentos Intermediarios/deficiencia , Proteínas de Filamentos Intermediarios/genética , Ratones , Microbiota , Mutación , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/genética , Transducción de Señal , Piel/inmunología , Piel/microbiología , Células Th2/inmunología , Linfopoyetina del Estroma Tímico , Interleucina-22
13.
FASEB J ; 30(2): 824-35, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26490658

RESUMEN

Induction of a type 2 cellular response in the white adipose tissue leads to weight loss and improves glucose homeostasis in obese animals. Injection of obese mice with recombinant helminth-derived Schistosoma mansoni egg-derived ω1 (ω1), a potent inducer of type 2 activation, improves metabolic status involving a mechanism reliant upon release of the type 2 initiator cytokine IL-33. IL-33 initiates the accumulation of group 2 innate lymphoid cells (ILC2s), eosinophils, and alternatively activated macrophages in the adipose tissue. IL-33 release from cells in the adipose tissue is mediated by the RNase activity of ω1; however, the ability of ω1 to improve metabolic status is reliant upon effective binding of ω1 to CD206. We demonstrate a novel mechanism for RNase-mediated release of IL-33 inducing ILC2-dependent improvements in the metabolic status of obese animals.


Asunto(s)
Antígenos Helmínticos/inmunología , Proteínas del Huevo/inmunología , Proteínas del Helminto/inmunología , Inmunidad Innata , Interleucina-33/inmunología , Linfocitos/inmunología , Ribonucleasas/inmunología , Schistosoma mansoni/inmunología , Animales , Eosinófilos/inmunología , Interleucina-33/genética , Lectinas Tipo C/inmunología , Activación de Macrófagos/genética , Macrófagos/inmunología , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Obesidad/inmunología , Receptores de Superficie Celular/inmunología , Schistosoma mansoni/enzimología
14.
Proc Natl Acad Sci U S A ; 111(1): 367-72, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24344271

RESUMEN

Disease conditions associated with pulmonary fibrosis are progressive and have a poor long-term prognosis with irreversible changes in airway architecture leading to marked morbidity and mortalities. Using murine models we demonstrate a role for interleukin (IL)-25 in the generation of pulmonary fibrosis. Mechanistically, we identify IL-13 release from type 2 innate lymphoid cells (ILC2) as sufficient to drive collagen deposition in the lungs of challenged mice and suggest this as a potential mechanism through which IL-25 is acting. Additionally, we demonstrate that in human idiopathic pulmonary fibrosis there is increased pulmonary expression of IL-25 and also observe a population ILC2 in the lungs of idiopathic pulmonary fibrosis patients. Collectively, we present an innate mechanism for the generation of pulmonary fibrosis, via IL-25 and ILC2, that occurs independently of T-cell-mediated antigen-specific immune responses. These results suggest the potential of therapeutically targeting IL-25 and ILC2 for the treatment of human fibrotic diseases.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-17/metabolismo , Interleucinas/metabolismo , Linfocitos/citología , Fibrosis Pulmonar/metabolismo , Anciano , Animales , Moléculas de Adhesión Celular/metabolismo , Colágeno/química , Colágeno/metabolismo , Femenino , Humanos , Fibrosis Pulmonar Idiopática/patología , Inmunidad Innata , Inflamación , Interleucina-13/metabolismo , Hígado/parasitología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fibrosis Pulmonar/patología , Schistosoma mansoni
15.
J Allergy Clin Immunol ; 137(2): 482-91, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26299987

RESUMEN

BACKGROUND: Atopic dermatitis (AD) is an inflammatory skin condition that can occur in early life, predisposing to asthma development in a phenomenon known as the atopic march. Although genetic and environmental factors are known to contribute to AD and asthma, the mechanisms underlying the atopic march remain poorly understood. Filaggrin loss-of-function mutations are a major genetic predisposer for the development of AD and progression to AD-associated asthma. OBJECTIVE: We sought to experimentally address whether filaggrin mutations in mice lead to the development of spontaneous eczematous inflammation and address the aberrant immunologic milieu arising in a mouse model of filaggrin deficiency. METHODS: Filaggrin mutant mice were generated on the proallergic BALB/c background, creating a novel model for the assessment of spontaneous AD-like inflammation. Independently recruited AD case collections were analyzed to define associations between filaggrin mutations and immunologic phenotypes. RESULTS: Filaggrin-deficient mice on a BALB/c background had profound spontaneous AD-like inflammation with progression to compromised pulmonary function with age, reflecting the atopic march in patients with AD. Strikingly, skin inflammation occurs independently of adaptive immunity and is associated with cutaneous expansion of IL-5-producing type 2 innate lymphoid cells. Furthermore, subjects with filaggrin mutations have an increased frequency of type 2 innate lymphoid cells in the skin in comparison with control subjects. CONCLUSION: This study provides new insights into our understanding of the atopic march, with innate immunity initiating dermatitis and the adaptive immunity required for subsequent development of compromised lung function.


Asunto(s)
Inmunidad Adaptativa , Dermatitis Atópica/complicaciones , Dermatitis Atópica/inmunología , Inmunidad Innata , Neumonía/etiología , Animales , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Proteínas Filagrina , Proteínas de Filamentos Intermediarios/deficiencia , Proteínas de Filamentos Intermediarios/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Neumonía/patología , Piel/inmunología , Piel/metabolismo , Piel/patología
16.
Eur J Immunol ; 45(6): 1842-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25763771

RESUMEN

B cells have been described as having the capacity to regulate cellular immune responses and suppress inflammatory processes. One such regulatory B-cell population is defined as IL-10-producing CD19(+) CD1d(hi) cells. Previous work has identified an expansion of these cells in mice infected with the helminth, Schistosoma mansoni. Here, microarray analysis of CD19(+) CD1d(hi) B cells from mice infected with S. mansoni demonstrated significantly increased Tlr7 expression, while CD19(+) CD1d(hi) B cells from uninfected mice also demonstrated elevated Tlr7 expression. Using IL-10 reporter, Il10(-/-) and Tlr7(-/-) mice, we formally demonstrate that TLR7 ligation of CD19(+) CD1d(hi) B cells increases their capacity to produce IL-10. In a mouse model of allergic lung inflammation, the adoptive transfer of TLR7-elicited CD19(+) CD1d(hi) B cells reduced airway inflammation and associated airway hyperresponsiveness. Using DEREG mice to deplete FoxP3(+) T regulatory cells in allergen-sensitized mice, we show that that TLR7-elicited CD19(+) CD1d(hi) B cells suppress airway hyperresponsiveness via a T regulatory cell dependent mechanism. These studies identify that TLR7 stimulation leads to the expansion of IL-10-producing CD19(+) CD1d(hi) B cells, which can suppress allergic lung inflammation via T regulatory cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Antígenos CD19/metabolismo , Antígenos CD1d/metabolismo , Modelos Animales de Enfermedad , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Interleucina-10/biosíntesis , Ratones , Ratones Noqueados , Ovalbúmina/efectos adversos , Neumonía/parasitología , Unión Proteica , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/parasitología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Regulación hacia Arriba
17.
Proc Natl Acad Sci U S A ; 110(46): 18490-5, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24145445

RESUMEN

Hypoxia is a prominent feature of chronically inflamed tissues. Oxygen-sensing hydroxylases control transcriptional adaptation to hypoxia through the regulation of hypoxia-inducible factor (HIF) and nuclear factor κB (NF-κB), both of which can regulate the inflammatory response. Furthermore, pharmacologic hydroxylase inhibitors reduce inflammation in multiple animal models. However, the underlying mechanism(s) linking hydroxylase activity to inflammatory signaling remains unclear. IL-1ß, a major proinflammatory cytokine that regulates NF-κB, is associated with multiple inflammatory pathologies. We demonstrate that a combination of prolyl hydroxylase 1 and factor inhibiting HIF hydroxylase isoforms regulates IL-1ß-induced NF-κB at the level of (or downstream of) the tumor necrosis factor receptor-associated factor 6 complex. Multiple proteins of the distal IL-1ß-signaling pathway are subject to hydroxylation and form complexes with either prolyl hydroxylase 1 or factor inhibiting HIF. Thus, we hypothesize that hydroxylases regulate IL-1ß signaling and subsequent inflammatory gene expression. Furthermore, hydroxylase inhibition represents a unique approach to the inhibition of IL-1ß-dependent inflammatory signaling.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipoxia/fisiopatología , Inflamación/fisiopatología , Oxigenasas de Función Mixta/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Western Blotting , Células HeLa , Humanos , Hidroxilación , Hipoxia/metabolismo , Inmunoprecipitación , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Luciferasas , Espectrometría de Masas , Prolil Hidroxilasas/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
18.
Mol Cancer ; 14: 45, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25889301

RESUMEN

BACKGROUND: The VEGF pathway has become an important therapeutic target in lung cancer, where VEGF has long been established as a potent pro-angiogenic growth factor expressed by many types of tumors. While Bevacizumab (Avastin) has proven successful in increasing the objective tumor response rate and in prolonging progression and overall survival in patients with NSCLC, the survival benefit is however relatively short and the majority of patients eventually relapse. The current use of tyrosine kinase inhibitors alone and in combination with chemotherapy has been underwhelming, highlighting an urgent need for new targeted therapies. In this study, we examined the mechanisms of VEGF-mediated survival in NSCLC cells and the role of the Neuropilin receptors in this process. METHODS: NSCLC cells were screened for expression of VEGF and its receptors. The effects of recombinant VEGF and its blockade on lung tumor cell proliferation and cell cycle were examined. Phosphorylation of Akt and Erk1/2 proteins was examined by high content analysis and confocal microscopy. The effects of silencing VEGF on cell proliferation and survival signaling were also assessed. A Neuropilin-1 stable-transfected cell line was generated. Cell growth characteristics in addition to pAkt and pErk1/2 signaling were studied in response to VEGF and its blockade. Tumor growth studies were carried out in nude mice following subcutaneous injection of NP1 over-expressing cells. RESULTS: Inhibition of the VEGF pathway with anti-VEGF and anti-VEGFR-2 antibodies or siRNA to VEGF, NP1 and NP2 resulted in growth inhibition of NP1 positive tumor cell lines associated with down-regulation of PI3K and MAPK kinase signaling. Stable transfection of NP1 negative cells with NP1 induced proliferation in vitro, which was further enhanced by exogenous VEGF. In vivo, NP1 over-expressing cells significantly increased tumor growth in xenografts compared to controls. CONCLUSIONS: Our data demonstrate that VEGF is an autocrine growth factor in NSCLC signaling, at least in part, through NP1. Targeting this VEGF receptor may offer potential as a novel therapeutic approach and also support the evaluation of the role of NP1 as a biomarker predicting sensitivity or resistance to VEGF and VEGFR-targeted therapies in the clinical arena.


Asunto(s)
Proteína C-Reactiva/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas del Tejido Nervioso/genética , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo/genética , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Fosfatidilinositol 3-Quinasas/genética , Receptores de Factores de Crecimiento Endotelial Vascular/genética
19.
J Immunol ; 191(11): 5349-53, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24166975

RESUMEN

The cellular composition of visceral adipose tissue (VAT) and release of cytokines by such cells within VAT has been implicated in regulating obesity and metabolic homeostasis. We show the importance of IL-25-responsive innate cells, which release the Th2 cytokine IL-13, in regulating weight and glucose homeostasis in mouse models of diet-induced obesity. Treating obese mice with IL-25 induces weight loss and improves glucose tolerance, and is associated with increased infiltration of innate lymphoid type 2 cells (ILC2), type I and type II NKT cells, eosinophils, and alternatively activated macrophages into the VAT. By depleting ILC2 in obese Rag1(-/-) mice, we observe exacerbated weight gain and glucose intolerance. Conversely, transferring ILC2 or type I or type II NKT cells into obese mice induces transient weight loss and stabilizes glucose homeostasis. Our data identify a mechanism whereby IL-25 eliciting IL-13-producing innate cells regulates inflammation in adipose tissue and prevents diet-induced obesity.


Asunto(s)
Interleucinas/inmunología , Grasa Intraabdominal/inmunología , Subgrupos Linfocitarios/inmunología , Células T Asesinas Naturales/inmunología , Obesidad/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Movimiento Celular , Glucosa/inmunología , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Homeostasis , Inmunidad Innata , Inmunomodulación , Interleucina-13/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética
20.
Haematologica ; 99(1): 185-93, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23872307

RESUMEN

Vitamin K-dependent proteases generated in response to vascular injury and infection enable fibrin clot formation, but also trigger distinct immuno-regulatory signaling pathways on myeloid cells. Factor Xa, a protease crucial for blood coagulation, also induces protease-activated, receptor-dependent cell signaling. Factor Xa can bind both monocytes and macrophages, but whether factor Xa-dependent signaling stimulates or suppresses myeloid cell cytokine production in response to Toll-like receptor activation is not known. In this study, exposure to factor Xa significantly impaired pro-inflammatory cytokine production from lipopolysaccharide-treated peripheral blood mononuclear cells, THP-1 monocytic cells and murine macrophages. Furthermore, factor Xa inhibited nuclear factor-kappa B activation in THP-1 reporter cells, requiring phosphatidylinositide 3-kinase activity for its anti-inflammatory effect. Active-site blockade, γ-carboxyglutamic acid domain truncation and a peptide mimic of the factor Xa inter-epidermal growth factor-like region prevented factor Xa inhibition of lipopolysaccharide-induced tumor necrosis factor-α release. In addition, factor Xa anti-inflammatory activity was markedly attenuated by the presence of an antagonist of protease-activated receptor 2, but not protease-activated receptor 1. The key role of protease-activated receptor 2 in eliciting factor Xa-dependent anti-inflammatory signaling on macrophages was further underscored by the inability of factor Xa to mediate inhibition of tumor necrosis factor-α and interleukin-6 release from murine bone marrow-derived protease-activated receptor 2-deficient macrophages. We also show for the first time that, in addition to protease-activated receptor 2, factor Xa requires a receptor-associated protein-sensitive low-density lipoprotein receptor to inhibit lipopolysaccharide-induced cytokine production. Collectively, the findings of this study support a novel function for factor Xa as an endogenous, receptor-associated protein-sensitive, protease-activated receptor 2-dependent regulator of myeloid cell pro-inflammatory cytokine production.


Asunto(s)
Citocinas/biosíntesis , Factor Xa/metabolismo , Mediadores de Inflamación/metabolismo , Células Mieloides/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal , Androstadienos/farmacología , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Factor Xa/química , Humanos , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Células Mieloides/inmunología , FN-kappa B/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptor PAR-2/genética , Transducción de Señal/efectos de los fármacos , Wortmanina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda