Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Respir Res ; 22(1): 207, 2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34271910

RESUMEN

BACKGROUND: Alpha-1 antitrypsin (AAT) is a major serine protease inhibitor. AAT deficiency (AATD) is a genetic disorder characterized by early-onset severe emphysema. In well-selected AATD patients, therapy with plasma-derived AAT (pAAT), "augmentation therapy", provides modest clinical improvement but is perceived as cumbersome with weekly intravenous infusions. Using mouse models of emphysema, we compared the effects of a recombinant AAT-IgG1 Fc-fusion protein (AAT-Fc), which is expected to have a longer half-life following infusion, to those of pAAT. METHODS: In an elastase model of emphysema, mice received a single intratracheal instillation of porcine pancreatic elastase (PPE) or human leucocyte elastase (hLE). AAT-Fc, pAAT, or vehicle was administered intraperitoneally 1 day prior to or 3 weeks following elastase instillation. Lung function and histology assessments were performed at 7 and 32 days after elastase instillation. In a cigarette smoke (CS) model of emphysema, mice were exposed to CS daily, 5 days a week, for 6 months and AAT-Fc, pAAT, or vehicle were administered every 10 days during the last 3 months of CS exposure. Assessments were performed 3 days after the last CS exposure. Immune responses to lung elastin peptide (EP) and the effects of AAT-Fc or pAAT treatment on dendritic cell (DC) function were determined ex vivo. RESULTS: Both elastase instillation and CS exposure triggered emphysema-like alveolar enlargement, increased lung compliance, and increased markers of inflammation compared to controls. Administration of AAT-Fc either prior to or following elastase instillation or during CS exposure provided greater protection than pAAT against alveolar enlargement, lung dysfunction, and airway inflammation. When challenged ex vivo with EP, spleen mononuclear cells from elastase-exposed mice exhibited dose-dependent production of IFNγ and IL-17, suggesting immune reactivity. In co-culture experiments with splenic CD4+ T cells isolated from elastase-exposed mice, AAT-Fc treatment prior to EP-priming of bone marrow-derived dendritic cells inhibited the production of IFNγ and IL-17. CONCLUSIONS: Compared to pAAT, AAT-Fc more effectively prevented or attenuated elastase- and CS-induced models of emphysema. These effects were associated with immunomodulatory effects on DC activity. AAT-Fc may provide a therapeutic option to individuals with AATD- and CS-induced emphysema.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Exposición por Inhalación/efectos adversos , Elastasa Pancreática/toxicidad , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Humo/efectos adversos , alfa 1-Antitripsina/administración & dosificación , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Elastasa Pancreática/administración & dosificación , Enfisema Pulmonar/inmunología , Porcinos , Nicotiana
2.
J Allergy Clin Immunol ; 145(3): 933-946.e4, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31626843

RESUMEN

BACKGROUND: Inducible CD4+CD25+ regulatory T (iTreg) cells can become pathogenic effector cells, enhancing lung allergic responses. OBJECTIVE: We aimed to define the underlying cellular and molecular pathways activated by TGF-ß, which determine the suppressor or enhancing activities of iTreg cells. METHODS: Sensitized wild-type and CD8-deficient (CD8-/-) mice were challenged with allergen. Isolated CD4+CD25- T cells were activated by using anti-CD3/anti-CD28. To generate suppressor iTreg cells, cells were then differentiated in the presence of TGF-ß, whereas IL-17-producing effector T cells were additionally exposed to IL-6. After TGF-ß, Smad3 and TGF-ß-activated kinase 1 (TAK1) kinase levels were monitored. The consequences of inhibiting either kinase were determined in vitro and after transfer into CD8-/- recipients. Quantitative PCR and chromatin immunoprecipitation were used to monitor gene expression and histone modifications at the retinoic acid-related orphan receptor γt (Rorγt) locus. RESULTS: In wild-type mice, iTreg cells suppressed lung allergic responses linked to Smad3-dependent forkhead box P3 (Foxp3) expression and IL-10 production. In the presence of IL-6, iTreg cells converted to TH17 cells, mediating a neutrophil-dependent enhancement of lung allergic responses in CD8-/- mice. Conversion was regulated by TAK1. Inhibition or silencing of TAK1 prevented expression of Rorγt and TH17 differentiation through histone modifications of Rorγt; Foxp3 expression and iTreg cell-mediated suppression remained intact. In the same cell, TGF-ß induced coexpression of Smad3 and TAK1 proteins; in the presence of IL-6, expression of Smad3 and Foxp3 but not TAK1 decreased. CONCLUSION: TGF-ß regulates iTreg cell outcomes through 2 distinct signal transduction pathways: one Smad3 dependent and the other TAK1 dependent. The balance of these pathways has important implications in TH17-mediated autoimmune diseases and neutrophil-dependent asthma.


Asunto(s)
Quinasas Quinasa Quinasa PAM/inmunología , Hipersensibilidad Respiratoria/inmunología , Proteína smad3/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Diferenciación Celular/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/inmunología
3.
Immunol Rev ; 278(1): 63-86, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28658551

RESUMEN

Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.


Asunto(s)
Hipersensibilidad/inmunología , Inmunomodulación , Activación de Linfocitos/inmunología , Neumonía/inmunología , Linfocitos T/inmunología , Animales , Diferenciación Celular , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Hipersensibilidad/genética , Hipersensibilidad/metabolismo , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Neumonía/genética , Neumonía/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
4.
J Immunol ; 200(4): 1261-1269, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29352000

RESUMEN

Mesenchymal stem cells (MSC) exert immune modulatory properties and previous studies demonstrated suppressive effects of MSC treatment in animal models of allergic airway inflammation. However, the underlying mechanisms have not been fully elucidated. We studied the role of MSC in immune activation and subsequent recruitment of monocytes in suppressing airway hyperresponsiveness and airway inflammation using a mouse model of allergic airway inflammation. MSC administration prior to or after allergen challenge inhibited the development of airway inflammation in allergen-sensitized mice. This was accompanied by an influx of CCR2-positive monocytes, which were localized around injected MSC in the lungs. Notably, IL-10-producing monocytes and/or macrophages were also increased in the lungs. Systemic administration of liposomal clodronate or a CCR2 antagonist significantly prevented the suppressive effects of MSC. Activation of MSC by IFN-γ leading to the upregulation of CCL2 expression was essential for the suppressive effects, as administration of wild-type MSC into IFN-γ-deficient recipients, or IFN-γ receptor-deficient or CCL2-deficient MSC into wild-type mice failed to suppress airway inflammation. These results suggest that MSC activation by IFN-γ, followed by increased expression of CCL2 and recruitment of monocytes to the lungs, is essential for suppression by MSC in allergen-induced airway hyperresponsiveness and airway inflammation.


Asunto(s)
Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Receptores CCR2/inmunología , Hipersensibilidad Respiratoria/inmunología , Animales , Movimiento Celular/inmunología , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/biosíntesis , Hipersensibilidad Respiratoria/metabolismo
5.
J Allergy Clin Immunol ; 143(6): 2026-2037.e7, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30660639

RESUMEN

BACKGROUND: CD8+ type 2 cytotoxic T (TC2) cells undergo transcriptional reprogramming to IL-13 production in the presence of IL-4 to become potent, steroid-insensitive, pathogenic effector cells in asthmatic patients and in mice in a model of experimental asthma. However, no studies have described the effects of hypoxia exposure on TC2 cell differentiation. OBJECTIVE: We determined the effects of hypoxia exposure on IL-13-producing CD8+ TC2 cells. METHODS: CD8+ transgenic OT-1 cells differentiated with IL-2 and IL-4 (TC2 cells) were exposed to normoxia (21% oxygen) or hypoxia (3% oxygen), and IL-13 production in vitro was monitored. After differentiation under these conditions, cells were adoptively transferred into CD8-deficient mice, and lung allergic responses, including airway hyperresponsiveness to inhaled methacholine, were assessed. The effects of pharmacologic inhibitors of hypoxia-inducible factor (HIF) 1α and HIF-2α were determined, as were responses in HIF-1α-deficient OT-1 cells. RESULTS: Under hypoxic conditioning, CD8+ TC2 cell differentiation was significantly enhanced, with increased numbers of IL-13+ T cells and increased production of IL-13 in vitro. Adoptive transfer of TC2 cells differentiated under hypoxic conditioning restored lung allergic responses in sensitized and challenged CD8-deficient recipients to a greater degree than seen in recipients of TC2 cells differentiated under normoxic conditioning. Pharmacologic inhibition of HIF-1α or genetic manipulation to reduce HIF-1α expression reduced the hypoxia-enhanced differentiation of TC2 cells, IL-13 production, and the capacity of transferred cells to restore lung allergic responses in vivo. IL-4-dependent, hypoxia-mediated increases in HIF-1α and TC2 cell differentiation were shown to be mediated through activation of Janus kinase 1/3 and GATA-3. CONCLUSIONS: Hypoxia enhances CD8+ TC2 cell-dependent airway hyperresponsiveness and inflammation through HIF-1α activation. These findings coupled with the known insensitivity of CD8+ T cells to corticosteroids suggests that activation of the IL-4-HIF-1α-IL-13 axis might play a role in the development of steroid-refractory asthma.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Hipoxia/inmunología , Inflamación/inmunología , Hipersensibilidad Respiratoria/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Broncoconstrictores , Células Cultivadas , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Pulmón/inmunología , Pulmón/fisiopatología , Cloruro de Metacolina , Ratones Transgénicos , Hipersensibilidad Respiratoria/fisiopatología
6.
J Allergy Clin Immunol ; 141(2): 659-670.e2, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28479331

RESUMEN

BACKGROUND: Regulatory T (Treg) cells play an essential role in the maintenance of immune homeostasis in allergic diseases. OBJECTIVES: We sought to define the mechanisms underlying induction of tolerance to peanut protein and prevention of the development of peanut allergy. METHODS: High or low doses of peanut extract were administered to pups every day for 2 weeks before peanut sensitization and challenge. After challenge, symptoms, Treg cell numbers, and forkhead box protein 3 (Foxp3), TH2 and TH17 cytokine, and Tgfß expression in mesenteric lymph node (MLN) CD4+ T cells and jejunum were monitored. Treg cell suppressive activity and Foxp3 methylation in MLN CD4+ T cells were assayed. RESULTS: Feeding high but not low doses of peanut before sensitization induced tolerance, as demonstrated by prevention of diarrhea and peanut-specific IgE responses, increases in the percentage of CD4+CD25+FoxP3+ cells in MLNs, and Foxp3 mRNA and protein expression in CD4+ cells from MLNs or jejunum. Feeding high doses of peanut before sensitization decreased percentages of CD3+CD4+IL-13+ and CD3+CD4+IL-17+ cells in MLNs and decreased Il13 and Il17a and increased Tgfß mRNA expression in the jejunum; numbers of CD103+ dendritic cells in MLNs were significantly increased. Treg cell suppression was shown to be antigen specific. Foxp3 methylation was increased in peanut extract-sensitized and challenged mice, whereas in tolerized mice levels were significantly reduced. CONCLUSIONS: Feeding high doses of peanut to pups induced tolerance to peanut protein. Foxp3 demethylation was associated with tolerance induction, indicating that Treg cells play an important role in the regulation of peanut sensitivity and maintenance of immune homeostasis.


Asunto(s)
Arachis/química , Factores de Transcripción Forkhead/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Yeyuno/inmunología , Hipersensibilidad al Cacahuete , Extractos Vegetales/farmacología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Citocinas/inmunología , Desmetilación/efectos de los fármacos , Modelos Animales de Enfermedad , Yeyuno/patología , Ratones , Ratones Endogámicos BALB C , Hipersensibilidad al Cacahuete/inmunología , Hipersensibilidad al Cacahuete/patología , Hipersensibilidad al Cacahuete/prevención & control , Extractos Vegetales/química , Extractos Vegetales/inmunología , Linfocitos T Colaboradores-Inductores/patología
7.
Proc Natl Acad Sci U S A ; 112(16): 5111-6, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25820174

RESUMEN

Silencing of interleukin-32 (IL-32) in a differentiated human promonocytic cell line impairs killing of Mycobacterium tuberculosis (MTB) but the role of IL-32 in vivo against MTB remains unknown. To study the effects of IL-32 in vivo, a transgenic mouse was generated in which the human IL-32γ gene is expressed using the surfactant protein C promoter (SPC-IL-32γTg). Wild-type and SPC-IL-32γTg mice were infected with a low-dose aerosol of a hypervirulent strain of MTB (W-Beijing HN878). At 30 and 60 d after infection, the transgenic mice had 66% and 85% fewer MTB in the lungs and 49% and 68% fewer MTB in the spleens, respectively; the transgenic mice also exhibited greater survival. Increased numbers of host-protective innate and adaptive immune cells were present in SPC-IL-32γTg mice, including tumor necrosis factor-alpha (TNFα) positive lung macrophages and dendritic cells, and IFN-gamma (IFNγ) and TNFα positive CD4(+) and CD8(+) T cells in the lungs and mediastinal lymph nodes. Alveolar macrophages from transgenic mice infected with MTB ex vivo had reduced bacterial burden and increased colocalization of green fluorescent protein-labeled MTB with lysosomes. Furthermore, mouse macrophages made to express IL-32γ but not the splice variant IL-32ß were better able to limit MTB growth than macrophages capable of producing both. The lungs of patients with tuberculosis showed increased IL-32 expression, particularly in macrophages of granulomas and airway epithelial cells but also B cells and T cells. We conclude that IL-32γ enhances host immunity to MTB.


Asunto(s)
Interleucinas/metabolismo , Mycobacterium tuberculosis/patogenicidad , Tuberculosis/inmunología , Tuberculosis/prevención & control , Inmunidad Adaptativa/inmunología , Animales , Antígenos Ly/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Humanos , Inmunidad Innata/inmunología , Interferón gamma , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macrófagos Alveolares/inmunología , Ratones Transgénicos , Mutación/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Sitios de Empalme de ARN/genética , Linfocitos T Reguladores/inmunología , Transfección , Transgenes , Tuberculosis/microbiología , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia/inmunología
8.
J Allergy Clin Immunol ; 139(4): 1331-1342, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27542981

RESUMEN

BACKGROUND: Regulatory T cells attenuate development of asthma in wild-type (WT) mice, with both naturally occurring regulatory T (nTreg) cells and inducible regulatory T (iTreg) cells exhibiting suppressive activity. When transferred into CD8-deficient (CD8-/-) recipients, both cell types enhanced development of allergen-induced airway hyperresponsiveness. OBJECTIVE: We sought to determine whether the pathways leading to enhancement of lung allergic responses by transferred nTreg and iTreg cells differed. METHODS: nTreg cells (CD4+CD25+) were isolated from WT mice and iTreg cells were generated from WT CD4+CD25- T cells after activation in the presence of TGF-ß and transferred into sensitized CD8-/- recipients before challenge. Development of airway hyperresponsiveness, cytokine levels, and airway inflammation were monitored. RESULTS: Transfer of nTreg cells enhanced lung allergic responses, as did transfer of iTreg cells. Although anti-IL-13 reduced nTreg cell-mediated enhancement, it was ineffective in iTreg cell-mediated enhancement; conversely, anti-IL-17, but not anti-IL-13, attenuated the enhancement by iTreg cells. Recovered iTreg cells from the lungs of CD8-/- recipients were capable of IL-17 production and expressed high levels of signature genes of the TH17 pathway, RORγt and Il17, whereas reduced expression of the Treg cell key transcription factor forkhead box p3 (Foxp3) was observed. In vitro exogenous IL-6-induced IL-17 production in iTreg cells, and in vivo conversion of transferred iTreg cells was dependent on recipient IL-6. CONCLUSIONS: iTreg cells, similar to nTreg cells, exhibit functional plasticity and can be converted from suppressor cells to pathogenic effector cells, enhancing lung allergic responses, but these effects were mediated through different pathways.


Asunto(s)
Asma/inmunología , Hiperreactividad Bronquial/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Hipersensibilidad/inmunología , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
J Immunol ; 193(5): 2238-47, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070841

RESUMEN

Glucocorticoid-induced TNFR family-related protein (GITR)-mediated activation of JNK was shown to regulate the suppressive activity of CD4(+)CD25(+) naturally occurring T regulatory cells (nTregs) in wild-type (WT) hosts. In this study, CD4(+)CD25(+) T cells were shown to be capable of becoming pathogenic effector cells in sensitized and challenged CD8(-/-) recipient mice. Only GITR-expressing CD4(+)CD25(+) T cells, but neither GITR knocked-in CD4(+)CD25(-) T cells nor GITR-silenced CD4(+)CD25(+) T cells, enhanced development of lung allergic responses. Inhibition of JNK in WT nTregs or nTregs from GITR(-/-)and JNK2(-/-) mice failed to enhance lung allergic responses in sensitized and challenged CD8(-/-) recipient mice. The failure to enhance responses was associated with increased bronchoalveolar lavage fluid levels of IL-10 and TGF-ß and decreased levels of IL-5, IL-6, and IL-13. In contrast, nTregs from JNK1(-/-) mice, similar to WT nTregs, were fully effective in enhancing responses. Thus, GITR stimulation of nTregs and signaling through JNK2, but not JNK1, triggered the loss of regulatory function while concomitantly gaining pathogenic CD4(+) T effector cell function responsible for exacerbating asthma-like immunopathology.


Asunto(s)
Asma/inmunología , Pulmón/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 9 Activada por Mitógenos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Asma/genética , Asma/patología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Pulmón/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/inmunología , Proteína Quinasa 9 Activada por Mitógenos/genética , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
10.
J Immunol ; 193(10): 4757-60, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25381356

RESUMEN

Changes made in the 8th edition of the Guide for the Care and Use of Laboratory Animals included new recommendations for the amount of space for breeding female mice. Adopting the new recommendations required, in essence, the elimination of trio breeding practices for all institutions. Both public opinion and published data did not readily support the new recommendations. In response, the National Jewish Health Institutional Animal Care and Use Committee established a program to directly compare the effects of breeding format on mouse pup survival and growth. Our study showed an overall parity between trio and pairwise breeding formats on the survival and growth of the litters, suggesting that the housing recommendations for breeding female mice as stated in the current Guide for the Care and Use of Laboratory Animals should be reconsidered.


Asunto(s)
Cruzamiento/métodos , Vivienda para Animales/ética , Animales , Autoinmunidad , Peso Corporal , Cruzamiento/legislación & jurisprudencia , Femenino , Guías como Asunto , Vivienda para Animales/legislación & jurisprudencia , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Embarazo
11.
Proc Natl Acad Sci U S A ; 110(20): 8152-7, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23630275

RESUMEN

Allergic asthma is a heterogeneous inflammatory disorder of the airways characterized by chronic airway inflammation and airway hyperresponsiveness. Numbers of CD8(+)IL-13(+) T cells are increased in asthmatics and during the development of experimental asthma in mice. In an atopic environment rich in IL-4, these CD8(+) T cells mediate asthmatic responses, but the mechanisms regulating the conversion of CD8(+) effector T cells from IFN-γ- to pathogenic IL-13-producing effector cells that contribute to an asthma phenotype have not been defined. Here, we show that cholesterol side-chain cleavage P450 enzyme, Cyp11a1, is a key regulator of CD8(+) T-cell conversion. Expression of the gene, protein, and enzymatic activity of Cyp11a1 were markedly increased in CD8(+) T cells differentiated in the presence of IL-2 plus IL-4 compared with cells differentiated in IL-2 alone. Inhibition of Cyp11a1 enzymatic activity with aminoglutethimide or reduction in the expression of Cyp11a1 using short hairpin RNA prevented the IL-4-induced conversion of IFN-γ- to IL-13-producing cells without affecting expression of the lineage-specific transcription factors T-box expressed in T cells (T-bet) or GATA binding protein 3 (GATA3). Adoptive transfer of aminoglutethimide-treated CD8(+) T cells into sensitized and challenged CD8-deficient recipients failed to restore airway hyperresponsiveness and inflammation. We demonstrate that Cyp11a1 controls the phenotypic conversion of CD8(+) T cells from IFN-γ to IL-13 production, linking steroidogenesis in CD8(+) T cells, a nonclassical steroidogenic tissue, to a proallergic differentiation pathway.


Asunto(s)
Linfocitos T CD8-positivos/citología , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Enfermedades Pulmonares/metabolismo , Animales , Asma/metabolismo , Líquido del Lavado Bronquioalveolar , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Ensayo de Inmunoadsorción Enzimática/métodos , Hipersensibilidad/metabolismo , Interferón gamma/metabolismo , Interleucina-13/metabolismo , Interleucina-2/farmacología , Interleucina-4/farmacología , Ratones , Ratones Endogámicos C57BL , Pregnenolona/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
12.
J Allergy Clin Immunol ; 135(2): 451-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25312762

RESUMEN

BACKGROUND: Eosinophils accumulate at the site of allergic inflammation and are critical effector cells in allergic diseases. Recent studies have also suggested a role for eosinophils in the resolution of inflammation. OBJECTIVE: To determine the role of eosinophils in the resolution phase of the response to repeated allergen challenge. METHODS: Eosinophil-deficient (PHIL) and wild-type (WT) littermates were sensitized and challenged to ovalbumin (OVA) 7 or 11 times. Airway inflammation, airway hyperresponsiveness (AHR) to inhaled methacholine, bronchoalveolar lavage (BAL) cytokine levels, and lung histology were monitored. Intracellular cytokine levels in BAL leukocytes were analyzed by flow cytometry. Groups of OVA-sensitized PHIL mice received bone marrow from WT or IL-10(-/-) donors 30 days before the OVA challenge. RESULTS: PHIL and WT mice developed similar levels of AHR and numbers of leukocytes and cytokine levels in BAL fluid after OVA sensitization and 7 airway challenges; no eosinophils were detected in the PHIL mice. Unlike WT mice, sensitized PHIL mice maintained AHR, lung inflammation, and increased levels of IL-4, IL-5, and IL-13 in BAL fluid after 11 challenges whereas IL-10 and TGF-ß levels were decreased. Restoration of eosinophil numbers after injection of bone marrow from WT but not IL-10-deficient mice restored levels of IL-10 and TGF-ß in BAL fluid as well as suppressed AHR and inflammation. Intracellular staining of BAL leukocytes revealed the capacity of eosinophils to produce IL-10. CONCLUSIONS: After repeated allergen challenge, eosinophils appeared not essential for the development of AHR and lung inflammation but contributed to the resolution of AHR and inflammation by producing IL-10.


Asunto(s)
Alérgenos/inmunología , Eosinófilos/inmunología , Hipersensibilidad/inmunología , Pulmón/inmunología , Alérgenos/administración & dosificación , Animales , Trasplante de Médula Ósea , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Femenino , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Recuento de Leucocitos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , Ovalbúmina/inmunología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo
13.
J Immunol ; 190(2): 539-48, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23241885

RESUMEN

Histamine H(4) receptor (H(4)R)-deficient mice (H(4)R(-/-)), H(4)R antagonist-treated wild-type (WT) mice, and WT mice depleted of basophils failed to develop early (EPR) or late phase (LPR) nasal responses following allergen sensitization and challenge. Basophil transfer from WT but not H(4)R(-/-) mice restored the EPR and LPR in H(4)R(-/-) mice. Following passive sensitization with OVA-specific IgE, FcεRI(-/-) recipients of WT basophils plus OVA and histamine developed an EPR and LPR. OVA-IgE passively sensitized FcεRI(-/-) recipients of H(4)R(-/-) basophils and OVA and histamine challenge failed to develop an EPR or LPR, and basophils were not detected in nasal tissue. In contrast, recipients of basophils from IL-13(-/-) and IL-4(-/-)/IL-13(-/-) mice developed an EPR but not an LPR. These results demonstrate the development of allergic rhinitis proceeded in two distinct stages: histamine release from FcεRI-activated mast cells, followed by histamine-mediated recruitment of H(4)R-expressing basophils to the nasal cavity and activation through FcεRI.


Asunto(s)
Basófilos/inmunología , Mastocitos/inmunología , Receptores Acoplados a Proteínas G/inmunología , Receptores Histamínicos/inmunología , Receptores de IgE/inmunología , Rinitis Alérgica Perenne/inmunología , Traslado Adoptivo , Animales , Basófilos/efectos de los fármacos , Basófilos/metabolismo , Femenino , Antagonistas de los Receptores Histamínicos/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , Ratones Noqueados , Modelos Inmunológicos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos/genética , Receptores Histamínicos H4 , Receptores de IgE/deficiencia , Receptores de IgE/genética , Rinitis Alérgica , Rinitis Alérgica Perenne/genética , Rinitis Alérgica Perenne/terapia
14.
J Immunol ; 190(8): 4056-65, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509358

RESUMEN

The functional plasticity of CD8(+) T cells in an atopic environment, encompassing a spectrum from IFN-γ- to IL-13-producing cells, is pivotal in the development of allergic airway hyperresponsiveness and inflammation, and yet remains mechanistically undefined. We demonstrate that CD8(+) T cell IL-13 induction proceeded through a series of distinct IL-4/GATA3-regulated stages characterized by gene expression and epigenetic changes. In vivo, CD8(+) T cells exposed to an environment rich in IL-4 displayed epigenetic changes at the GATA3 and IL-13 promoter indicative of transcriptional activation and IL-13 production. In vitro, IL-4 triggered the stepwise molecular conversion of CD8(+) T cells from IFN-γ to IL-13 production. During the initial stage, IL-4 suppressed T-bet and induced GATA3 expression, characterized by enhanced activating histone modifications and RNA polymerase II (Pol II) recruitment to the GATA3 locus. Notably, recruitment of GATA3 and RNA Pol II to the IL-13 promoter was also detected at this initial stage. However, enhanced IL-13 transcription only occurred at a later stage after TCR stimulation, indicating that IL-4-induced GATA3 recruitment poises the IL-13 locus for TCR-mediated transcription. Thus, both in vivo and in vitro, an atopic (IL-4) environment poises CD8(+) T cells via stepwise epigenetic and phenotypic mechanisms for pathogenic conversion to IL-13 production, which is ultimately triggered via an allergen-mediated TCR stimulus.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Epigénesis Genética/inmunología , Inmunofenotipificación , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Animales , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Inmunofenotipificación/métodos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Hipersensibilidad Respiratoria/metabolismo
15.
J Allergy Clin Immunol ; 133(4): 1162-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24365136

RESUMEN

BACKGROUND: Janus kinases (JAKs) are regulators of signaling through cytokine receptors. The importance of JAK1/3 signaling on TH2 differentiation and development of lung allergic responses has not been investigated. OBJECTIVE: We sought to examine a selective JAK1/3 inhibitor (R256) on differentiation of TH subsets in vitro and on development of ovalbumin (OVA)-induced airway hyperresponsiveness (AHR) and inflammation in an experimental model of asthma. METHODS: A selective JAK1/3 inhibitor was used to assay the importance of this pathway on induction of TH1, TH2, and TH17 differentiation in vitro. In vivo, the effects of inhibiting JAK1/3 signaling were examined by administering the inhibitor during the sensitization or allergen challenge phases in the primary challenge model or just before provocative challenge in the secondary challenge model. Airway inflammation and AHR were examined after the last airway challenge. RESULTS: In vitro, R256 inhibited differentiation of TH2 but not TH1 or TH17 cells, which was associated with downregulation of signal transducer and activator of transcription (STAT) 6 and STAT5 phosphorylation. However, once polarized, TH2 cells were unaffected by the inhibitor. In vivo, R256 administered during the OVA sensitization phase prevented the development of AHR, airway eosinophilia, mucus hypersecretion, and TH2 cytokine production without changes in TH1 and TH17 cytokine levels, indicating that selective blockade of TH2 differentiation was critical. Inhibitor administration after OVA sensitization but during the challenge phases in the primary or secondary challenge models similarly suppressed AHR, airway eosinophilia, and mucus hypersecretion without any reduction in TH2 cytokine production, suggesting the inhibitory effects were downstream of TH2 cytokine receptor signaling pathways. CONCLUSIONS: Targeting the TH2-dependent JAK/STAT activation pathway represents a novel therapeutic approach for the treatment of asthma.


Asunto(s)
Diferenciación Celular , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Transducción de Señal , Células Th2/citología , Células Th2/metabolismo , Alérgenos/administración & dosificación , Alérgenos/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacología , Hipersensibilidad Respiratoria/patología , Transducción de Señal/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Células Th2/efectos de los fármacos , Células Th2/inmunología
16.
Am J Respir Cell Mol Biol ; 51(1): 143-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24521403

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illnesses in infants worldwide. Both RSV-G and RSV-F glycoproteins play pathogenic roles during infection with RSV. The objective of this study was to compare the effects of anti-RSV-G and anti-RSV-F monoclonal antibodies (mAbs) on airway hyperresponsiveness (AHR) and inflammation after primary or secondary RSV infection in mice. In the primary infection model, mice were infected with RSV at 6 weeks of age. Anti-RSV-G or anti-RSV-F mAbs were administered 24 hours before infection or Day +2 postinfection. In a secondary infection model, mice were infected (primary) with RSV at 1 week (neonate) and reinfected (secondary) 5 weeks later. Anti-RSV-G and anti-RSV-F mAbs were administered 24 hours before the primary infection. Both mAbs had comparable effects in preventing airway responses after primary RSV infection. When given 2 days after infection, anti-RSV-G-treated mice showed significantly decreased AHR and airway inflammation, which persisted in anti-RSV-F-treated mice. In the reinfection model, anti-RSV-G but not anti-RSV-F administered during primary RSV infection in neonates resulted in decreased AHR, eosinophilia, and IL-13 but increased levels of IFN-γ in bronchoalveolar lavage on reinfection. These results support the use of anti-RSV-G in the prevention and treatment of RSV-induced disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Bronquiolitis Viral/prevención & control , Inflamación/prevención & control , Hipersensibilidad Respiratoria/prevención & control , Infecciones por Virus Sincitial Respiratorio/prevención & control , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Bronquiolitis Viral/etiología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inflamación/etiología , Ratones , Ratones Endogámicos BALB C , Hipersensibilidad Respiratoria/etiología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/patogenicidad
17.
Ann Allergy Asthma Immunol ; 112(3): 211-216.e1, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24428972

RESUMEN

BACKGROUND: Numbers of CD8(+) T cells expressing the leukotriene B4 (LTB4) receptor, BLT1, have been correlated with asthma severity. OBJECTIVE: To examine the activation and numbers of BLT1-expressing peripheral blood CD4(+) and CD8(+) T cells from patients with steroid-sensitive (SS) and steroid-resistant (SR) asthma. METHODS: CD4(+) and CD8(+) T cells isolated from peripheral blood of healthy human subjects and patients with SS and SR asthma were stimulated in culture with anti-CD3/anti-CD28 followed by analysis of BLT1 surface expression and cytokine production. Activation of CD8(+) T cells after ligation of BLT1 by LTB4 was monitored by changes in intracellular Ca(2+) concentrations. RESULTS: The number of BLT1-expressing cells was larger in patients with asthma than in controls and larger on activated CD8(+) than on CD4(+) T cells. Addition of LTB4 to activated CD8(+) T cells resulted in increases in intracellular Ca(2+) concentrations. Expansion of activated CD4(+) T cells, unlike CD8(+) T cells, was significantly decreased in the presence of corticosteroid. In patients with SS asthma, numbers of BLT1-expressing CD8(+) T cells were lower in the presence of corticosteroid, unlike in those with SR asthma in whom cell expansion was maintained. Levels of interleukin-13 were highest in cultured CD8(+) T cells, whereas interleukin-10 levels were higher in CD4(+) T cells from controls and patients with SS asthma. Interferon-γ levels were lowest in patients with SR asthma. CONCLUSION: Differences in BLT1 expression, steroid sensitivity, and cytokine production were demonstrated in T lymphocytes from patients with SS and SR asthma. The LTB4-BLT1 pathway in CD8(+) cells may play an important role in asthma and serve as an important target in the treatment of patients with SR asthma.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Dexametasona/farmacología , Receptores de Leucotrieno B4/metabolismo , Corticoesteroides/farmacología , Adulto , Anticuerpos Monoclonales/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Calcio/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/química , Femenino , Humanos , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Activación de Linfocitos/inmunología , Masculino
18.
J Immunol ; 188(2): 661-7, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22174452

RESUMEN

Activation of the alternative pathway of complement plays a critical role in the development of allergen-induced airway hyperresponsiveness (AHR) and inflammation in mice. Endogenous factor H, a potent inhibitor of the alternative pathway, is increased in the airways of sensitized and challenged mice, but its role in regulating inflammation or AHR has been unknown. We found that blocking the tissue-binding function of factor H with a competitive antagonist increased complement activation and tissue inflammation after allergen challenge of sensitized mice. Conversely, administration of a fusion protein that contains the iC3b/C3d binding region of complement receptor 2 linked to the inhibitory region of factor H, a molecule directly targeting complement-activating surfaces, protected mice in both primary and secondary challenge models of AHR and lung inflammation. Thus, although endogenous factor H does play a role in limiting the development of AHR, strategies to deliver the complement-regulatory region of factor H specifically to the site of inflammation provide greater protection than that afforded by endogenous regulators. Such an agent may be an effective therapy for the treatment of asthma.


Asunto(s)
Alérgenos/administración & dosificación , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Factor H de Complemento/fisiología , Vía Alternativa del Complemento/inmunología , Mediadores de Inflamación/fisiología , Alérgenos/inmunología , Animales , Asma/inmunología , Asma/metabolismo , Asma/patología , Hiperreactividad Bronquial/metabolismo , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología
19.
J Immunol ; 189(7): 3404-10, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22933632

RESUMEN

Heat shock proteins (HSPs), produced in response to stress, are suppressive in disease models. We previously showed that Mycobacterium leprae HSP65 prevented development of airway hyperresponsiveness and inflammation in mice. Our goal in this study was to define the mechanism responsible for the suppressive effects of HSP. In one in vivo approach, BALB/c mice were sensitized to OVA, followed by primary OVA challenges. Several weeks later, HSP65 was administered prior to a single, provocative secondary challenge. In a second in vivo approach, the secondary challenge was replaced by intratracheal instillation of allergen-pulsed bone marrow-derived dendritic cells (BMDCs). The in vitro effects of HSP65 on BMDCs were examined in coculture experiments with CD4(+) T cells. In vivo, HSP65 prevented the development of airway hyperresponsiveness and inflammation. Additionally, Th1 cytokine levels in bronchoalveolar lavage fluid were increased. In vitro, HSP65 induced Notch receptor ligand Delta1 expression on BMDCs, and HSP65-treated BMDCs skewed CD4(+) T cells to Th1 cytokine production. Thus, HSP65-induced effects on allergen-induced airway hyperresponsiveness and inflammation were associated with increased Delta1 expression on dendritic cells, modulation of dendritic cell function, and CD4(+) Th1 cytokine production.


Asunto(s)
Proteínas Bacterianas/fisiología , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/prevención & control , Chaperonina 60/fisiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Inflamación/prevención & control , Mycobacterium leprae/inmunología , Animales , Hiperreactividad Bronquial/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/biosíntesis , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Células TH1/inmunología , Células TH1/microbiología , Células TH1/patología
20.
J Biol Chem ; 287(21): 17100-17108, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22461627

RESUMEN

Naturally occurring Foxp3(+)CD4(+)CD25(+) T regulatory cell (nTreg)-mediated suppression of lung allergic responses is abrogated following ligation of glucocorticoid-induced tumor necrosis receptor (GITR) family-related protein. In vitro stimulation of nTregs with GITR ligand increased phosphorylation of c-Jun N-terminal kinase (JNK) but not extracellular signal-regulated protein kinase (ERK) or p38 MAPK. SP600125, a known JNK inhibitor, prevented GITR-mediated phosphorylation of JNK. Activation of JNK was associated with increases in the upstream mitogen-activated protein kinase kinase 7 (MKK7) and the downstream transcription factor NF-κß. Phosphorylated c-Jun (p-c-Jun), indicative of the activation of JNK, was detected in the immunoprecipitates of nTregs from wild-type but not JNK- or GITR-deficient mice. Treatment with an inhibitor of JNK phosphorylation resulted in complete reversal of all GITR-induced changes in nTreg phenotype and function, with full restoration of suppression of in vivo lung allergic responses and in vitro proliferation of activated CD4(+)CD25(-) T cells. Thus, regulation of JNK phosphorylation plays a central role in T regulatory cell function with therapeutic implications for the treatment of asthma and autoimmune diseases.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Tolerancia Inmunológica/fisiología , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Linfocitos T Reguladores/metabolismo , Animales , Antracenos/farmacología , Asma/genética , Asma/inmunología , Asma/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/inmunología , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/inmunología , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosforilación/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T Reguladores/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA