Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Immunity ; 54(7): 1463-1477.e11, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34115964

RESUMEN

Acute respiratory distress syndrome (ARDS), an inflammatory condition with high mortality rates, is common in severe COVID-19, whose risk is reduced by metformin rather than other anti-diabetic medications. Detecting of inflammasome assembly in post-mortem COVID-19 lungs, we asked whether and how metformin inhibits inflammasome activation while exerting its anti-inflammatory effect. We show that metformin inhibited NLRP3 inflammasome activation and interleukin (IL)-1ß production in cultured and alveolar macrophages along with inflammasome-independent IL-6 secretion, thus attenuating lipopolysaccharide (LPS)- and SARS-CoV-2-induced ARDS. By targeting electron transport chain complex 1 and independently of AMP-activated protein kinase (AMPK) or NF-κB, metformin blocked LPS-induced and ATP-dependent mitochondrial (mt) DNA synthesis and generation of oxidized mtDNA, an NLRP3 ligand. Myeloid-specific ablation of LPS-induced cytidine monophosphate kinase 2 (CMPK2), which is rate limiting for mtDNA synthesis, reduced ARDS severity without a direct effect on IL-6. Thus, inhibition of ATP and mtDNA synthesis is sufficient for ARDS amelioration.


Asunto(s)
Adenosina Trifosfato/metabolismo , ADN Mitocondrial/biosíntesis , Inflamasomas/efectos de los fármacos , Metformina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/prevención & control , Animales , COVID-19/metabolismo , COVID-19/prevención & control , Citocinas/genética , Citocinas/metabolismo , ADN Mitocondrial/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Metformina/uso terapéutico , Ratones , Nucleósido-Fosfato Quinasa/metabolismo , Neumonía/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2/patogenicidad
2.
Immunity ; 42(3): 538-51, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25769613

RESUMEN

Allergic asthma is caused by Th2-cell-type cytokines in response to allergen exposure. Type 2 innate lymphoid cells (ILC2s) are a newly identified subset of immune cells that, along with Th2 cells, contribute to the pathogenesis of asthma by producing copious amounts of IL-5 and IL-13, which cause eosinophilia and airway hyperreactivity (AHR), a cardinal feature of asthma. ILC2s express ICOS, a T cell costimulatory molecule with a currently unknown function. Here we showed that a lack of ICOS on murine ILC2s and blocking the ICOS:ICOS-ligand interaction in human ILC2s reduced AHR and lung inflammation. ILC2s expressed both ICOS and ICOS-ligand, and the ICOS:ICOS-ligand interaction promoted cytokine production and survival in ILC2s through STAT5 signaling. Thus, ICOS:ICOS-ligand signaling pathway is critically involved in ILC2 function and homeostasis.


Asunto(s)
Asma/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Linfocitos/inmunología , Animales , Asma/genética , Asma/patología , Femenino , Regulación de la Expresión Génica , Homeostasis , Humanos , Inmunidad Innata , Ligando Coestimulador de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-33 , Interleucina-5/genética , Interleucina-5/inmunología , Interleucinas/genética , Interleucinas/inmunología , Linfocitos/patología , Ratones Transgénicos , Sistema Respiratorio/inmunología , Sistema Respiratorio/patología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Transducción de Señal
3.
J Allergy Clin Immunol ; 151(2): 526-538.e8, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35963455

RESUMEN

BACKGROUND: Neutrophilic asthma is associated with disease severity and corticosteroid insensitivity. Novel therapies are required to manage this life-threatening asthma phenotype. Programmed cell death protein-1 (PD-1) is a key homeostatic modulator of the immune response for T-cell effector functions. OBJECTIVE: We sought to investigate the role of PD-1 in the regulation of acute neutrophilic inflammation in a murine model of airway hyperreactivity (AHR). METHODS: House dust mite was used to induce and compare neutrophilic AHR in wild-type and PD-1 knockout mice. Then, the therapeutic potential of a human PD-1 agonist was tested in a humanized mouse model in which the PD-1 extracellular domain is entirely humanized. Single-cell RNA sequencing and flow cytometry were mainly used to investigate molecular and cellular mechanisms. RESULTS: PD-1 was highly induced on pulmonary T cells in our inflammatory model. PD-1 deficiency was associated with an increased neutrophilic AHR and high recruitment of inflammatory cells to the lungs. Consistently, PD-1 agonist treatment dampened AHR, decreased neutrophil recruitment, and modulated cytokine production in a humanized PD-1 mouse model. Mechanistically, we demonstrated at the transcriptional and protein levels that the inhibitory effect of PD-1 agonist is associated with the reprogramming of pulmonary effector T cells that showed decreased number and activation. CONCLUSIONS: PD-1 agonist treatment is efficient in dampening neutrophilic AHR and lung inflammation in a preclinical humanized mouse model.


Asunto(s)
Asma , Receptor de Muerte Celular Programada 1 , Humanos , Animales , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Pulmón , Células Th2 , Modelos Animales de Enfermedad
4.
Allergy ; 78(7): 1922-1933, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36929161

RESUMEN

BACKGROUND: The impact of exposure to air pollutants, such as fine particulate matter (PM), on the immune system and its consequences on pediatric asthma, are not well understood. We investigated whether ambient levels of fine PM with aerodynamic diameter ≤2.5 microns (PM2.5 ) are associated with alterations in circulating monocytes in children with or without asthma. METHODS: Monocyte phenotyping was performed by cytometry time-of-flight (CyTOF). Cytokines were measured using cytometric bead array and Luminex assay. ChIP-Seq was utilized to address histone modifications in monocytes. RESULTS: Increased exposure to ambient PM2.5 was linked to specific monocyte subtypes, particularly in children with asthma. Mechanistically, we hypothesized that innate trained immunity is evoked by a primary exposure to fine PM and accounts for an enhanced inflammatory response after secondary stimulation in vitro. We determined that the trained immunity was induced in circulating monocytes by fine particulate pollutants, and it was characterized by the upregulation of proinflammatory mediators, such as TNF, IL-6, and IL-8, upon stimulation with house dust mite or lipopolysaccharide. This phenotype was epigenetically controlled by enhanced H3K27ac marks in circulating monocytes. CONCLUSION: The specific alterations of monocytes after ambient pollution exposure suggest a possible prognostic immune signature for pediatric asthma, and pollution-induced trained immunity may provide a potential therapeutic target for asthmatic children living in areas with increased air pollution.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Asma , Humanos , Material Particulado/efectos adversos , Monocitos , Inmunidad Entrenada , Contaminantes Atmosféricos/efectos adversos , Contaminantes Atmosféricos/análisis , Asma/etiología , Asma/inducido químicamente , Contaminación del Aire/efectos adversos
5.
J Allergy Clin Immunol ; 147(4): 1281-1295.e5, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32905799

RESUMEN

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are the dominant innate lymphoid cell population in the lungs at steady state, and their release of type 2 cytokines is a central driver in responding eosinophil infiltration and increased airway hyperreactivity. Our laboratory has identified a unique subset of ILC2s in the lungs that actively produce IL-10 (ILC210s). OBJECTIVE: Our aim was to characterize the effector functions of ILC210s in the development and pathology of allergic asthma. METHODS: IL-4-stimulated ILC210s were isolated to evaluate cytokine secretion, transcription factor signaling, metabolic dependence, and effector functions in vitro. ILC210s were also adoptively transferred into Rag2-/-γc-/- mice, which were then challenged with IL-33 and assessed for airway hyperreactivity and lung inflammation. RESULTS: We have determined that the transcription factors cMaf and Blimp-1 regulate IL-10 expression in ILC210s. Strikingly, our results demonstrate that ILC210s can utilize both autocrine and paracrine signaling to suppress proinflammatory ILC2 effector functions in vitro. Further, this subset dampens airway hyperreactivity and significantly reduces lung inflammation in vivo. Interestingly, ILC210s demonstrated a metabolic dependency on the glycolytic pathway for IL-10 production, shifting from the fatty acid oxidation pathway conventionally utilized for proinflammatory effector functions. CONCLUSION: These findings provide an important and previously unrecognized role of ILC210s in diseases associated with ILC2s such as allergic lung inflammation and asthma. They also provide new insights into the metabolism dependency of proinflammatory and anti-inflammatory ILC2 phenotypes.


Asunto(s)
Asma/inmunología , Hiperreactividad Bronquial/inmunología , Interleucina-10/inmunología , Linfocitos/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Proteínas Proto-Oncogénicas c-maf/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
J Allergy Clin Immunol ; 145(2): 502-517.e5, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31738991

RESUMEN

BACKGROUND: Allergic asthma is a chronic inflammatory disorder characterized by airway hyperreactivity (AHR) and driven by TH2 cytokine production. Group 2 innate lymphoid cells (ILC2s) secrete high amounts of TH2 cytokines and contribute to the development of AHR. Autophagy is a cellular degradation pathway that recycles cytoplasmic content. However, the role of autophagy in ILC2s remains to be fully elucidated. OBJECTIVE: We characterized the effects of autophagy deficiency on ILC2 effector functions and metabolic balance. METHODS: ILC2s from autophagy-deficient mice were isolated to evaluate proliferation, apoptosis, cytokine secretion, gene expression and cell metabolism. Also, autophagy-deficient ILC2s were adoptively transferred into Rag-/-GC-/- mice, which were then challenged with IL-33 and assessed for AHR and lung inflammation. RESULTS: We demonstrate that autophagy is extensively used by activated ILC2s to maintain their homeostasis and effector functions. Deletion of the critical autophagy gene autophagy-related 5 (Atg5) resulted in decreased cytokine secretion and increased apoptosis. Moreover, lack of autophagy among ILC2s impaired their ability to use fatty acid oxidation and strikingly promoted glycolysis, as evidenced by our transcriptomic and metabolite analyses. This shift of fuel dependency led to impaired homeostasis and TH2 cytokine production, thus inhibiting the development of ILC2-mediated AHR. Notably, this metabolic reprogramming was also associated with an accumulation of dysfunctional mitochondria, producing excessive reactive oxygen species. CONCLUSION: These findings provide new insights into the metabolic profile of ILC2s and suggest that modulation of fuel dependency by autophagy is a potentially new therapeutic approach to target ILC2-dependent inflammation.


Asunto(s)
Autofagia/inmunología , Homeostasis/inmunología , Inmunidad Innata/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Ratones , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo
7.
J Allergy Clin Immunol ; 141(3): 893-905.e6, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28579374

RESUMEN

BACKGROUND: Allergic asthma is a prevalent inflammatory disease of the airways caused by dysregulated immune balance in the lungs with incompletely understood pathogenesis. The recently identified type 2 innate lymphoid cells (ILC2s) play significant roles in the pathogenesis of asthma. Although ILC2-activating factors have been identified, the mechanisms that suppress ILC2s remain largely unknown. Plasmacytoid dendritic cells (pDCs) are important in antiviral immunity and in maintaining tolerance to inert antigens. OBJECTIVE: We sought to address the role of pDCs in regulating ILC2 function and ILC2-mediated airway hyperreactivity (AHR) and lung inflammation. METHODS: We used several murine models, including BDCA-2-diphtheria toxin receptor (DTR) transgenic and IFN-α receptor 1-deficient mice, as well as purified primary ILC2s, to reach our objective. We extended and validated our findings to human ILC2s. RESULTS: We show that activation of pDCs through Toll-like receptor 7/8 suppresses ILC2-mediated AHR and airway inflammation and that depletion of pDCs reverses this suppression. We further show that pDCs suppress cytokine production and the proliferation rate while increasing the apoptosis rate of ILC2s through IFN-α production. Transcriptomic analysis of both human and murine ILC2s confirms the activation of regulatory pathways in ILC2s by IFN-α. CONCLUSION: Activation of pDCs alleviates AHR and airway inflammation by suppressing ILC2 function and survival. Our findings reveal a novel regulatory pathway in ILC2-mediated pulmonary inflammation with important clinical implications.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Células Plasmáticas/inmunología , Animales , Asma/genética , Asma/patología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Células Plasmáticas/patología
8.
Immunity ; 30(6): 875-87, 2009 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-19501000

RESUMEN

NLRP3 nucleates the inflammasome, a protein complex responsible for cleavage of prointerleukin-1beta (IL-1beta) to its active form. Mutations in the NLRP3 gene cause the autoinflammatory disease spectrum cryopyrin-associated periodic syndromes (CAPS). The central role of IL-1beta in CAPS is supported by the response to IL-1-targeted therapy. We developed two Nlrp3 mutant knockin mouse strains to model CAPS to examine the role of other inflammatory mediators and adaptive immune responses in an innate immune-driven disease. These mice had systemic inflammation and poor growth, similar to some human CAPS patients, and demonstrated early mortality, primarily mediated by myeloid cells. Mating these mutant mice to various gene mutant backgrounds showed that the mouse disease phenotype required an intact inflammasome, was only partially dependent on IL-1beta, and was independent of T cells. These data suggest that CAPS are true inflammasome-mediated diseases and provide insight for more common inflammatory disorders.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Inmunidad Innata/genética , Inflamación/genética , Interleucina-1beta/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Inmunidad Activa , Inflamación/inmunología , Interleucina-18 , Interleucina-1beta/inmunología , Ratones , Ratones Mutantes , Mutación/genética , Mutación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
J Allergy Clin Immunol ; 139(5): 1468-1477.e2, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27717665

RESUMEN

BACKGROUND: Atopic diseases, including asthma, exacerbate type 2 immune responses and involve a number of immune cell types, including regulatory T (Treg) cells and the emerging type 2 innate lymphoid cells (ILC2s). Although ILC2s are potent producers of type 2 cytokines, the regulation of ILC2 activation and function is not well understood. OBJECTIVE: In the present study, for the first time, we evaluate how Treg cells interact with pulmonary ILC2s and control their function. METHODS: ILC2s and Treg cells were evaluated by using in vitro suppression assays, cell-contact assays, and gene expression panels. Also, human ILC2s and Treg cells were adoptively transferred into NOD SCID γC-deficient mice, which were given isotype or anti-inducible T-cell costimulator ligand (ICOSL) antibodies and then challenged with IL-33 and assessed for airway hyperreactivity. RESULTS: We show that induced Treg cells, but not natural Treg cells, effectively suppress the production of the ILC2-driven proinflammatory cytokines IL-5 and IL-13 both in vitro and in vivo. Mechanistically, our data reveal the necessity of inducible T-cell costimulator (ICOS)-ICOS ligand cell contact for Treg cell-mediated ILC2 suppression alongside the suppressive cytokines TGF-ß and IL-10. Using a translational approach, we then demonstrate that human induced Treg cells suppress syngeneic human ILC2s through ICOSL to control airway inflammation in a humanized ILC2 mouse model. CONCLUSION: These findings suggest that peripheral expansion of induced Treg cells can serve as a promising therapeutic target against ILC2-dependent asthma.


Asunto(s)
Asma/inmunología , Citocinas/inmunología , Linfocitos/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos
10.
J Immunol ; 195(5): 2429-41, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26209626

RESUMEN

Lung epithelial cells are considered important sources of inflammatory molecules and extracellular matrix proteins that contribute to diseases such as asthma. Understanding the factors that stimulate epithelial cells may lead to new insights into controlling lung inflammation. This study sought to investigate the responsiveness of human lung epithelial cells to the TNF family molecules LIGHT and lymphotoxin αß (LTαß). Bronchial and alveolar epithelial cell lines, and primary human bronchial epithelial cells, were stimulated with LIGHT and LTαß, and expression of inflammatory cytokines and chemokines and markers of epithelial-mesenchymal transition and fibrosis/remodeling was measured. LTß receptor, the receptor shared by LIGHT and LTαß, was constitutively expressed on all epithelial cells. Correspondingly, LIGHT and LTαß strongly induced a limited but highly distinct set of inflammatory genes in all epithelial cells tested, namely the adhesion molecules ICAM-1 and VCAM-1; the chemokines CCL5, CCL20, CXCL1, CXCL3, CXCL5, and CXCL11; the cytokines IL-6, activin A and GM-CSF; and metalloproteinases matrix metalloproteinase-9 and a disintegrin and metalloproteinase domain-8. Importantly, induction of the majority of these inflammatory molecules was insensitive to the suppressive effects of the corticosteroid budesonide. LIGHT and LTαß also moderately downregulated E-cadherin, a protein associated with maintaining epithelial integrity, but did not significantly drive production of extracellular matrix proteins or α-smooth muscle actin. Thus, LIGHT and LTαß induce a distinct steroid-resistant inflammatory signature in airway epithelial cells via constitutively expressed LTß receptor. These findings support our prior murine studies that suggested the receptors for LIGHT and LTαß contribute to development of lung inflammation characteristic of asthma and idiopathic pulmonary fibrosis.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Glucocorticoides/farmacología , Mediadores de Inflamación/metabolismo , Heterotrímero de Linfotoxina alfa1 y beta2/farmacología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Bronquios/citología , Budesonida/farmacología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Resistencia a Medicamentos , Células Epiteliales/metabolismo , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Proc Natl Acad Sci U S A ; 111(33): 12163-8, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092323

RESUMEN

The RAR-related orphan receptor gamma t (RORγt) is a nuclear receptor required for generating IL-17-producing CD4(+) Th17 T cells, which are essential in host defense and may play key pathogenic roles in autoimmune diseases. Oxysterols elicit profound effects on immune and inflammatory responses as well as on cholesterol and lipid metabolism. Here, we describe the identification of several naturally occurring oxysterols as RORγt agonists. The most potent and selective activator for RORγt is 7ß, 27-dihydroxycholesterol (7ß, 27-OHC). We show that these oxysterols reverse the inhibitory effect of an RORγt antagonist, ursolic acid, in RORγ- or RORγt-dependent cell-based reporter assays. These ligands bind directly to recombinant RORγ ligand binding domain (LBD), promote recruitment of a coactivator peptide, and reduce binding of a corepressor peptide to RORγ LBD. In primary cells, 7ß, 27-OHC and 7α, 27-OHC enhance the differentiation of murine and human IL-17-producing Th17 cells in an RORγt-dependent manner. Importantly, we showed that Th17, but not Th1 cells, preferentially produce these two oxysterols. In vivo, administration of 7ß, 27-OHC in mice enhanced IL-17 production. Mice deficient in CYP27A1, a key enzyme in generating these oxysterols, showed significant reduction of IL-17-producing cells, including CD4(+) and γδ(+) T cells, similar to the deficiency observed in RORγt knockout mice. Our results reveal a previously unknown mechanism for selected oxysterols as immune modulators and a direct role for CYP27A1 in generating these RORγt agonist ligands, which we propose as RORγt endogenous ligands, driving both innate and adaptive IL-17-dependent immune responses.


Asunto(s)
Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Esteroles/farmacología , Células Th17/citología , Animales , Diferenciación Celular , Colestanotriol 26-Monooxigenasa/metabolismo , Interleucina-17/biosíntesis , Ligandos , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Esteroles/metabolismo
12.
J Allergy Clin Immunol ; 137(5): 1382-1389.e9, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26589586

RESUMEN

BACKGROUND: Neutrophilic corticosteroid-resistant asthma accounts for a significant proportion of asthma; however, little is known about the mechanisms that underlie the pathogenesis of the disease. OBJECTIVE: We sought to address the role of autophagy in lung inflammation and the pathogenesis of corticosteroid-resistant neutrophilic asthma. METHODS: We developed CD11c-specific autophagy-related gene 5 (Atg5)(-/-) mice and used several murine models to investigate the role of autophagy in asthmatic patients. RESULTS: For the first time, we found that deletion of the Atg5 gene specifically in CD11c(+) cells, which leads to impairment of the autophagy pathway, causes unprovoked spontaneous airway hyperreactivity and severe neutrophilic lung inflammation in mice. We found that severe lung inflammation impairs the autophagy pathway, particularly in pulmonary CD11c(+) cells in wild-type mice. We further found that adoptive transfer of Atg5(-/-), but not wild-type, bone marrow-derived dendritic cells augments lung inflammation with increased IL-17A levels in the lungs. Our data indicate that neutrophilic asthma in Atg5(-/-) mice is glucocorticoid resistant and IL-17A dependent. CONCLUSION: Our results suggest that lack of autophagy in pulmonary CD11c(+) cells induces neutrophilic airway inflammation and hyperreactivity.


Asunto(s)
Asma , Autofagia , Dexametasona/uso terapéutico , Resistencia a Medicamentos , Animales , Antiinflamatorios/uso terapéutico , Asma/tratamiento farmacológico , Asma/inmunología , Proteína 5 Relacionada con la Autofagia/genética , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Recuento de Células , Citocinas/inmunología , Femenino , Pulmón/citología , Pulmón/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Pyroglyphidae/inmunología
13.
Eur J Immunol ; 44(10): 3015-25, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25103720

RESUMEN

T-cell homeostasis preserves the numbers, the diversity and functional competence of different T-cell subsets that are required for adaptive immunity. Naïve CD4(+) T (TN ) cells are maintained in the periphery via the common γ-chain family cytokine IL-7 and weak antigenic signals. However, it is not clear how memory CD4(+) T-cell subsets are maintained in the periphery and which factors are responsible for the maintenance. To examine the homeostatic mechanisms, CFSE-labeled CD4(+) CD44(high) CD62L(low) effector memory T (TEM ) cells were transferred into sublethally-irradiated syngeneic C57BL/6 mice, and the systemic cell proliferative responses, which can be divided distinctively into fast and slow proliferations, were assessed by CFSE dye dilution. We found that the fast homeostatic proliferation of TEM cells was strictly regulated by both antigen and OX40 costimulatory signals and that the slow proliferation was dependent on IL-7. The simultaneous blockade of both OX40 and IL-7 signaling completely inhibited the both fast and slow proliferation. The antigen- and OX40-dependent fast proliferation preferentially expanded IL-17-producing helper T cells (Th17 cells). Thus, OX40 and IL-7 play synergistic, but distinct roles in the homeostatic proliferation of CD4(+) TEM cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Memoria Inmunológica/inmunología , Interleucina-7/inmunología , Receptores OX40/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/fisiología , Citometría de Flujo , Homeostasis/inmunología , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores OX40/metabolismo , Subgrupos de Linfocitos T/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(7): 2903-8, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282629

RESUMEN

NF-κB activation is essential for T-cell responses, and costimulatory molecules in the TNF receptor (TNFR) superfamily are viewed as a major source of this signal. Although the TNFR family recruits TNFR-associated factor (TRAF) molecules leading to IKKα/ß/γ activation, it is not clear whether simple binding of TRAFs explains why they are such strong activators of NF-κB and so important for T-cell immunity. We now show that one TNFR family member, OX40 (CD134), after ligation by OX40L, assembles a unique complex that not only contains TRAF2, RIP, and IKKα/ß/γ but also CARMA1, MALT1, BCL10, and PKC, molecules previously shown to regulate NF-κB activation through the T-cell receptor (TCR). The OX40 signalosome is formed in membrane microdomains irrespective of TCR engagement, and strongly promotes NF-κB activation only if CARMA1 and PKC are recruited. This NF-κB signal allows effector/memory T cells to survive when antigen is no longer available. Thus, by recruiting TCR-related intracellular molecules into the TRAF2 complex, OX40 provides the T cell with a high level of NF-κB activity needed for longevity.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Isoenzimas/metabolismo , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Receptores OX40/metabolismo , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Animales , Western Blotting , Proteínas Adaptadoras de Señalización CARD/inmunología , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Isoenzimas/inmunología , Ratones , Complejos Multiproteicos/inmunología , Proteína Quinasa C/inmunología , Proteína Quinasa C-theta , Receptores OX40/inmunología , Linfocitos T/inmunología , Factor 2 Asociado a Receptor de TNF/inmunología , Ultracentrifugación
15.
J Allergy Clin Immunol ; 131(4): 1048-57, 1057.e1-2, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23174661

RESUMEN

BACKGROUND: Asthma is defined as a chronic inflammatory disease of the airways; however, the underlying physiologic and immunologic processes are not fully understood. OBJECTIVE: The aim of this study was to determine whether TH9 cells develop in vivo in a model of chronic airway hyperreactivity (AHR) and what factors control this development. METHOD: We have developed a novel chronic allergen exposure model using the clinically relevant antigen Aspergillus fumigatus to determine the time kinetics of TH9 development in vivo. RESULTS: TH9 cells were detectable in the lungs after chronic allergen exposure. The number of TH9 cells directly correlated with the severity of AHR, and anti-IL-9 treatment decreased airway inflammation. Moreover, we have identified programmed cell death ligand (PD-L) 2 as a negative regulator of TH9 cell differentiation. Lack of PD-L2 was associated with significantly increased TGF-ß and IL-1α levels in the lungs, enhanced pulmonary TH9 differentiation, and higher morbidity in the sensitized mice. CONCLUSION: Our findings suggest that PD-L2 plays a pivotal role in the regulation of TH9 cell development in chronic AHR, providing novel strategies for modulating adaptive immunity during chronic allergic responses.


Asunto(s)
Hiperreactividad Bronquial/genética , Interleucina-9/inmunología , Pulmón/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Subgrupos de Linfocitos T/inmunología , Inmunidad Adaptativa , Alérgenos/inmunología , Animales , Anticuerpos/inmunología , Aspergillus fumigatus/inmunología , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Diferenciación Celular/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Interleucina-1alfa/inmunología , Pulmón/metabolismo , Pulmón/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Índice de Severidad de la Enfermedad , Subgrupos de Linfocitos T/patología , Factor de Crecimiento Transformador beta/inmunología
17.
Immunol Rev ; 229(1): 173-91, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19426222

RESUMEN

SUMMARY: OX40 (CD134) and its binding partner, OX40L (CD252), are members of the tumor necrosis factor receptor/tumor necrosis factor superfamily and are expressed on activated CD4(+) and CD8(+) T cells as well as on a number of other lymphoid and non-lymphoid cells. Costimulatory signals from OX40 to a conventional T cell promote division and survival, augmenting the clonal expansion of effector and memory populations as they are being generated to antigen. OX40 additionally suppresses the differentiation and activity of T-regulatory cells, further amplifying this process. OX40 and OX40L also regulate cytokine production from T cells, antigen-presenting cells, natural killer cells, and natural killer T cells, and modulate cytokine receptor signaling. In line with these important modulatory functions, OX40-OX40L interactions have been found to play a central role in the development of multiple inflammatory and autoimmune diseases, making them attractive candidates for intervention in the clinic. Conversely, stimulating OX40 has shown it to be a candidate for therapeutic immunization strategies for cancer and infectious disease. This review provides a broad overview of the biology of OX40 including the intracellular signals from OX40 that impact many aspects of immune function and have promoted OX40 as one of the most prominent costimulatory molecules known to control T cells.


Asunto(s)
Antígenos CD40/inmunología , Ligando de CD40/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Humanos , Inmunoterapia , Activación de Linfocitos/inmunología , Neoplasias/terapia , Transducción de Señal/inmunología
18.
Cell Rep ; 42(8): 112990, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37590140

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe clinical disorders that mainly develop from viral respiratory infections, sepsis, and chest injury. Antigen-presenting cells play a pivotal role in propagating uncontrolled inflammation and injury through the excess secretion of pro-inflammatory cytokines and recruitment of immune cells. Autophagy, a homeostatic process that involves the degradation of cellular components, is involved in many processes including lung inflammation. Here, we use a polyinosinic-polycytidylic acid (poly(I:C))-induced lung injury mouse model to mimic viral-induced ALI/ARDS and show that disruption of autophagy in macrophages exacerbates lung inflammation and injury, whereas autophagy induction attenuates this process. Therefore, induction of autophagy in macrophages can be a promising therapeutic strategy in ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Animales , Ratones , Células Presentadoras de Antígenos , Macrófagos , Autofagia , Poli I-C/farmacología
19.
STAR Protoc ; 4(3): 102391, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37405925

RESUMEN

Here, we present a protocol for isolating human hepatocytes and neural progenitor cells from normal and nonalcoholic steatohepatitis livers. We describe steps for perfusion for scaled-up liver cell isolation and optimization of chemical digestion to achieve maximal yield and cell viability. We then detail a liver cell cryopreservation and potential applications, such as the use of human liver cells as a tool to link experimental and translational research.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Células Cultivadas , Hepatocitos , Separación Celular/métodos
20.
Nat Commun ; 13(1): 1440, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301333

RESUMEN

There has been a global increase in rates of obesity with a parallel epidemic of non-alcoholic fatty liver disease (NAFLD). Autophagy is an essential mechanism involved in the degradation of cellular material and has an important function in the maintenance of liver homeostasis. Here, we explore the effect of Autophagy-related 5 (Atg5) deficiency in liver CD11c+ cells in mice fed HFD. When compared to control mice, Atg5-deficient CD11c+ mice exhibit increased glucose intolerance and decreased insulin sensitivity when fed HFD. This phenotype is associated with the development of NAFLD. We observe that IL-23 secretion is induced in hepatic CD11c+ myeloid cells following HFD feeding. We demonstrate that both therapeutic and preventative IL-23 blockade alleviates glucose intolerance, insulin resistance and protects against NAFLD development. This study provides insights into the function of autophagy and IL-23 production by hepatic CD11c+ cells in NAFLD pathogenesis and suggests potential therapeutic targets.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Animales , Autofagia , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/genética , Interleucina-23/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda