Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 78(5): 814-823, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32333837

RESUMEN

Metabolites have functions in the immune system independent of their conventional roles as sources or intermediates in biosynthesis and bioenergetics. We are still in the pioneering phase of gathering information about the functions of specific metabolites in immunoregulation. In this review, we cover succinate, itaconate, α-ketoglutarate, and lactate as examples. Each of these metabolites has a different story of how their immunoregulatory functions were discovered and how their roles in the complex process of inflammation were revealed. Parallels and interactions are emerging between metabolites and cytokines, well-known immunoregulators. We depict molecular mechanisms by which metabolites prime cellular and often physiological changes focusing on intra- and extra-cellular activities and signaling pathways. Possible therapeutic opportunities for immune and inflammatory diseases are emerging.


Asunto(s)
Ácidos Carboxílicos/inmunología , Ácidos Carboxílicos/metabolismo , Inmunidad/inmunología , Animales , Ciclo del Ácido Cítrico , Citocinas/metabolismo , Metabolismo Energético , Humanos , Inmunidad/fisiología , Inflamación/metabolismo , Ácidos Cetoglutáricos/inmunología , Ácidos Cetoglutáricos/metabolismo , Ácido Láctico/inmunología , Ácido Láctico/metabolismo , Transducción de Señal , Succinatos/inmunología , Succinatos/metabolismo , Ácido Succínico/inmunología , Ácido Succínico/metabolismo
2.
J Biol Chem ; : 107793, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39305962

RESUMEN

Ubiquitin-Specific Peptidases (USPs) are the main members of deubiquitinases (DUBs) that catalyze removing ubiquitin chains from target proteins, thereby modulating their half-life and function. Enzymatic activity of USP21 regulates protein degradation which is critical for maintaining cell homeostasis. USP21 determines the stability of oncogenic proteins and therefore is implicated in carcinogenesis. In this study, we investigated the effect of USP21 deletion on cancer cell metabolism. Transcriptomic and proteomic analysis of USP21 knockout HAP-1 cells revealed that endogenous USP21 is critical for the expression of genes and proteins involved in mitochondrial function. Additionally, we have found that deletion of USP21 reduced STAT3 activation and STAT3-dependent gene and protein expression in cancer cells. Genetic deletion of USP21 impaired mitochondrial respiration and disturbed ATP production. This resulted in cellular consequences such as inhibition of cell proliferation and migration. Presented results provide new insights into the biology of USP21, suggesting novel mechanisms for controlling STAT3 activity and mitochondrial function in tumor cells. Taken together, our findings indicate that targeting USP21 dysregulates the energy status of cancer cells offering new perspectives for anti-cancer therapy.

3.
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
4.
J Immunol ; 207(10): 2561-2569, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34635585

RESUMEN

PGs are important proinflammatory lipid mediators, the significance of which is highlighted by the widespread and efficacious use of nonsteroidal anti-inflammatory drugs in the treatment of inflammation. 4-Octyl itaconate (4-OI), a derivative of the Krebs cycle-derived metabolite itaconate, has recently garnered much interest as an anti-inflammatory agent. In this article, we show that 4-OI limits PG production in murine macrophages stimulated with the TLR1/2 ligand Pam3CSK4. This decrease in PG secretion is due to a robust suppression of cyclooxygenase 2 (COX2) expression by 4-OI, with both mRNA and protein levels decreased. Dimethyl fumarate, a fumarate derivative used in the treatment of multiple sclerosis, with properties similar to itaconate, replicated the phenotype observed with 4-OI. We also demonstrate that the decrease in COX2 expression and inhibition of downstream PG production occurs in an NRF2-independent manner. Our findings provide a new insight into the potential of 4-OI as an anti-inflammatory agent and also identifies a novel anti-inflammatory function of dimethyl fumarate.


Asunto(s)
Antiinflamatorios/farmacología , Dimetilfumarato/farmacología , Macrófagos/efectos de los fármacos , Prostaglandinas/metabolismo , Succinatos/farmacología , Animales , Ciclooxigenasa 2/biosíntesis , Humanos , Macrófagos/metabolismo , Ratones
5.
J Immunol ; 207(10): 2551-2560, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34635586

RESUMEN

The protozoan parasite Trypanosoma brucei is the causative agent of the neglected tropical disease human African trypanosomiasis, otherwise known as sleeping sickness. Trypanosomes have evolved many immune-evasion mechanisms to facilitate their own survival, as well as prolonging host survival to ensure completion of the parasitic life cycle. A key feature of the bloodstream form of T. brucei is the secretion of aromatic keto acids, which are metabolized from tryptophan. In this study, we describe an immunomodulatory role for one of these keto acids, indole-3-pyruvate (I3P). We demonstrate that I3P inhibits the production of PGs in activated macrophages. We also show that, despite the reduction in downstream PGs, I3P augments the expression of cyclooxygenase (COX2). This increase in COX2 expression is mediated in part via inhibition of PGs relieving a negative-feedback loop on COX2. Activation of the aryl hydrocarbon receptor also participates in this effect. However, the increase in COX2 expression is of little functionality, as we also provide evidence to suggest that I3P targets COX activity. This study therefore details an evasion strategy by which a trypanosome-secreted metabolite potently inhibits macrophage-derived PGs, which might promote host and trypanosome survival.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Indoles/metabolismo , Macrófagos/inmunología , Prostaglandinas/metabolismo , Tripanosomiasis Africana/inmunología , Animales , Humanos , Evasión Inmune/inmunología , Indoles/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Prostaglandinas/inmunología , Trypanosoma brucei brucei/inmunología , Trypanosoma brucei brucei/metabolismo , Tripanosomiasis Africana/metabolismo
6.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36902148

RESUMEN

Chitotriosidase (CHIT1) is an enzyme produced by macrophages that regulates their differentiation and polarization. Lung macrophages have been implicated in asthma development; therefore, we asked whether pharmacological inhibition of macrophage-specific CHIT1 would have beneficial effects in asthma, as it has been shown previously in other lung disorders. CHIT1 expression was evaluated in the lung tissues of deceased individuals with severe, uncontrolled, steroid-naïve asthma. OATD-01, a chitinase inhibitor, was tested in a 7-week-long house dust mite (HDM) murine model of chronic asthma characterized by accumulation of CHIT1-expressing macrophages. CHIT1 is a dominant chitinase activated in fibrotic areas of the lungs of individuals with fatal asthma. OATD-01 given in a therapeutic treatment regimen inhibited both inflammatory and airway remodeling features of asthma in the HDM model. These changes were accompanied by a significant and dose-dependent decrease in chitinolytic activity in BAL fluid and plasma, confirming in vivo target engagement. Both IL-13 expression and TGFß1 levels in BAL fluid were decreased and a significant reduction in subepithelial airway fibrosis and airway wall thickness was observed. These results suggest that pharmacological chitinase inhibition offers protection against the development of fibrotic airway remodeling in severe asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Quitinasas , Inhibidores de Proteínas Quinasas , Animales , Humanos , Ratones , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/patología , Asma/terapia , Quitinasas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Pulmón/metabolismo , Macrófagos/enzimología , Pyroglyphidae/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
7.
FASEB J ; 34(3): 4718-4731, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32030817

RESUMEN

Resident alveolar macrophages (AMs) suppress allergic inflammation in murine asthma models. Previously we reported that resident AMs can blunt inflammatory signaling in alveolar epithelial cells (ECs) by transcellular delivery of suppressor of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here we examined the role of vesicular SOCS3 secretion as a mechanism by which AMs restrain allergic inflammatory responses in airway ECs. Bronchoalveolar lavage fluid (BALF) levels of SOCS3 were reduced in asthmatics and in allergen-challenged mice. Ex vivo SOCS3 secretion was reduced in AMs from challenged mice and this defect was mimicked by exposing normal AMs to cytokines associated with allergic inflammation. Both AM-derived EVs and synthetic SOCS3 liposomes inhibited the activation of STAT3 and STAT6 as well as cytokine gene expression in ECs challenged with IL-4/IL-13 and house dust mite (HDM) extract. This suppressive effect of EVs was lost when they were obtained from AMs exposed to allergic inflammation-associated cytokines. Finally, inflammatory cell recruitment and cytokine generation in the lungs of OVA-challenged mice were attenuated by intrapulmonary pretreatment with SOCS3 liposomes. Overall, AM secretion of SOCS3 within EVs serves as a brake on airway EC responses during allergic inflammation, but is impaired in asthma. Synthetic liposomes encapsulating SOCS3 can rescue this defect and may serve as a framework for novel therapeutic approaches targeting airway inflammation.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Adolescente , Adulto , Anciano , Animales , Asma/inmunología , Asma/metabolismo , Western Blotting , Línea Celular , Polaridad Celular/fisiología , Femenino , Humanos , Interleucina-33/metabolismo , Interleucina-4/metabolismo , Liposomas/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteína 3 Supresora de la Señalización de Citocinas/genética , Adulto Joven
8.
Proc Natl Acad Sci U S A ; 115(36): E8460-E8468, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30127006

RESUMEN

A variety of innate immune responses and functions are dependent on time of day, and many inflammatory conditions are associated with dysfunctional molecular clocks within immune cells. However, the functional importance of these innate immune clocks has yet to be fully characterized. NRF2 plays a critical role in the innate immune system, limiting inflammation via reactive oxygen species (ROS) suppression and direct repression of the proinflammatory cytokines, IL-1ß and IL-6. Here we reveal that the core molecular clock protein, BMAL1, controls the mRNA expression of Nrf2 via direct E-box binding to its promoter to regulate its activity. Deletion of Bmal1 decreased the response of NRF2 to LPS challenge, resulting in a blunted antioxidant response and reduced synthesis of glutathione. ROS accumulation was increased in Bmal1-/- macrophages, facilitating accumulation of the hypoxic response protein, HIF-1α. Increased ROS and HIF-1α levels, as well as decreased activity of NRF2 in cells lacking BMAL1, resulted in increased production of the proinflammatory cytokine, IL-1ß. The excessive prooxidant and proinflammatory phenotype of Bmal1-/- macrophages was rescued by genetic and pharmacological activation of NRF2, or through addition of antioxidants. Our findings uncover a clear role for the molecular clock in regulating NRF2 in innate immune cells to control the inflammatory response. These findings provide insights into the pathology of inflammatory conditions, in which the molecular clock, oxidative stress, and IL-1ß are known to play a role.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Factores de Transcripción ARNTL/genética , Animales , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Interleucina-1beta/genética , Lipopolisacáridos/toxicidad , Macrófagos/patología , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo
9.
Circ Res ; 122(6): 855-863, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29437834

RESUMEN

RATIONALE: Diurnal mechanisms are central to regulating host responses. Recent studies uncovered a novel family of mediators termed as specialized proresolving mediators that terminate inflammation without interfering with the immune response. OBJECTIVE: Herein, we investigated the diurnal regulation of specialized proresolving mediators in humans and their role in controlling peripheral blood leukocyte and platelet activation. METHODS AND RESULTS: Using lipid mediator profiling and healthy volunteers, we found that plasma concentrations of n-3 docosapentaenoic acid-derived D-series resolvins (RvDn-3 DPA) were regulated in a diurnal manner. The production and regulation of these mediators was markedly altered in patients at risk of myocardial infarct. These changes were associated with decreased 5-lipoxygenase expression and activity, as well as increased systemic adenosine concentrations. We also found a significant negative correlation between plasma RvDn-3 DPA and markers of platelet, monocyte, and neutrophil activation, including CD63 and CD11b. Incubation of RvDn-3 DPA with peripheral blood from healthy volunteers and patients with cardiovascular disease significantly and dose-dependently decreased platelet and leukocyte activation. Furthermore, administration of RvD5n-3 DPA to ApoE-/- (apolipoprotein E deficient) mice significantly reduced platelet-leukocyte aggregates, vascular thromboxane B2 concentrations, and aortic lesions. CONCLUSIONS: These results demonstrate that peripheral blood RvDn-3 DPA are diurnally regulated in humans, and dysregulation in the production of these mediators may lead to cardiovascular disease.


Asunto(s)
Ritmo Circadiano , Ácidos Grasos Insaturados/sangre , Infarto del Miocardio/sangre , Adenosina/sangre , Animales , Plaquetas/metabolismo , Humanos , Inflamación/sangre , Leucocitos/metabolismo , Lipooxigenasa/sangre , Ratones , Tromboxano B2/sangre
10.
Am J Respir Crit Care Med ; 200(11): 1381-1391, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31454256

RESUMEN

Rationale: Cystic fibrosis (CF) pulmonary disease is characterized by chronic infection with Pseudomonas aeruginosa and sustained neutrophil-dominant inflammation. The lack of effective antiinflammatory therapies for people with CF (PWCF) represents a significant challenge.Objectives: To identify altered immunometabolism in the CF neutrophil and investigate the feasibility of specific inhibition of the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome as a CF antiinflammatory strategy in vivo.Methods: Key markers of increased aerobic glycolysis, known as a Warburg effect, including cytosolic PKM2 (pyruvate kinase M2), phosphorylated PKM2, succinate, HIF-1α (hypoxia-inducible factor-1α), lactate, and the IL-1ß precursor pro-IL-1ß, as well as caspase-1 activity and processing of pro-IL-1ß to IL-1ß by the NLRP3 inflammasome, were measured in neutrophils from blood and airway secretions from healthy control subjects (n = 12), PWCF (n = 16), and PWCF after double-lung transplantation (n = 6). The effects of specific inhibition of NLRP3 on airway inflammation and bacterial clearance in a murine CF model were subsequently assessed in vivo.Measurements and Main Results: CF neutrophils display increased aerobic glycolysis in the systemic circulation. This effect is driven by low-level endotoxemia, unaffected by CFTR (cystic fibrosis transmembrane conductance regulator) modulation, and resolves after transplant. The increased pro-IL-1ß produced is processed to its mature active form in the LPS-rich CF lung by the NLRP3 inflammasome via caspase-1. Specific NLRP3 inhibition in vivo with MCC950 inhibited IL-1ß in the lungs of CF mice (P < 0.0001), resulting in significantly reduced airway inflammation and improved Pseudomonas clearance (P < 0.0001).Conclusions: CF neutrophil immunometabolism is altered in response to inflammation. NLRP3 inflammasome inhibition may have an antiinflammatory and anti-infective role in CF.


Asunto(s)
Antiinflamatorios/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Furanos/uso terapéutico , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Animales , Biomarcadores/análisis , Líquido del Lavado Bronquioalveolar/química , Compuestos Heterocíclicos de 4 o más Anillos , Humanos , Indenos , Interleucina-1beta/análisis , Ratones , Neutrófilos/efectos de los fármacos , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/terapia , Sulfonas
11.
J Biol Chem ; 293(15): 5509-5521, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29463677

RESUMEN

Different immune activation states require distinct metabolic features and activities in immune cells. For instance, inhibition of fatty acid synthase (FASN), which catalyzes the synthesis of long-chain fatty acids, prevents the proinflammatory response in macrophages; however, the precise role of this enzyme in this response remains poorly defined. Consistent with previous studies, we found here that FASN is essential for lipopolysaccharide-induced, Toll-like receptor (TLR)-mediated macrophage activation. Interestingly, only agents that block FASN upstream of acetoacetyl-CoA synthesis, including the well-characterized FASN inhibitor C75, inhibited TLR4 signaling, while those acting downstream had no effect. We found that acetoacetyl-CoA could overcome C75's inhibitory effect, whereas other FASN metabolites, including palmitate, did not prevent C75-mediated inhibition. This suggested an unexpected role for acetoacetyl-CoA in inflammation that is independent of its role in palmitate synthesis. Our evidence further suggested that acetoacetyl-CoA arising from FASN activity promotes cholesterol production, indicating a surprising link between fatty acid synthesis and cholesterol synthesis. We further demonstrate that this process is required for TLR4 to enter lipid rafts and facilitate TLR4 signaling. In conclusion, we have uncovered an unexpected link between FASN and cholesterol synthesis that appears to be required for TLR signal transduction and proinflammatory macrophage activation.


Asunto(s)
Colesterol/biosíntesis , Acido Graso Sintasa Tipo I/metabolismo , Activación de Macrófagos , Macrófagos/enzimología , Transducción de Señal , Acilcoenzima A/metabolismo , Animales , Inflamación/enzimología , Ratones , Ácido Palmítico/metabolismo , Receptor Toll-Like 4/metabolismo
12.
J Immunol ; 198(9): 3558-3564, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28298525

RESUMEN

PGE2 has been shown to increase the transcription of pro-IL-1ß. However, recently it has been demonstrated that PGE2 can block the maturation of IL-1ß by inhibiting the NLRP3 inflammasome in macrophages. These apparently conflicting results have led us to reexamine the effect of PGE2 on IL-1ß production. We have found that in murine bone marrow-derived macrophages, PGE2 via the cAMP/protein kinase A pathway is potently inducing IL-1ß transcription, as well as boosting the ability of LPS to induce IL-1ß mRNA and pro-IL-1ß while inhibiting the production of TNF-α. This results in an increase in mature IL-1ß production in macrophages treated with ATP. We also examined the effect of endogenously produced PGE2 on IL-1ß production. By blocking PGE2 production with indomethacin, we made a striking finding that endogenous PGE2 is essential for LPS-induced pro-IL-1ß production, suggesting a positive feedback loop. The effect of endogenous PGE2 was mediated by EP2 receptor. In primary human monocytes, where LPS alone is sufficient to induce mature IL-1ß, PGE2 boosted LPS-induced IL-1ß production. PGE2 did not inhibit ATP-induced mature IL-1ß production in monocytes. Because PGE2 mediates the pyrogenic effect of IL-1ß, these effects might be especially relevant for the role of monocytes in the induction of fever. A positive feedback loop from IL-1ß and back to PGE2, which itself is induced by IL-1ß, is likely to be operating. Furthermore, fever might therefore occur in the absence of a septic shock response because of the inhibiting effect of PGE2 on TNF-α production.


Asunto(s)
Dinoprostona/metabolismo , Fiebre/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Monocitos/inmunología , Adenosina Trifosfato/farmacología , Animales , Células Cultivadas , Dinoprostona/antagonistas & inhibidores , Retroalimentación Fisiológica , Humanos , Indometacina/farmacología , Inflamasomas/metabolismo , Interleucina-1beta/genética , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
13.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L855-L860, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28336811

RESUMEN

Our body clock drives rhythms in the expression of genes that have a 24-h periodicity. The transcription factor BMAL1 is a crucial component of the molecular clock. A number of physiological processes, including immune function, are modulated by the circadian clock. Asthma, a disease with very strong clinical evidence demonstrating regulation by circadian variation, is of particular relevance to circadian control of immunity. Airway hypersensitivity and asthma attacks are more common at night in humans. The molecular basis for this is unknown, and there is no model of asthma in animals with genetic distortion of the molecular clock. We used mice lacking BMAL1 in myeloid cells (BMAL1-LysM-/-) to determine the role of BMAL1 in allergic asthma. Using the ovalbumin model of allergic asthma, we demonstrated markedly increased asthma features, such as increased lung inflammation, demonstrated by drastically higher numbers of eosinophils and increased IL-5 levels in the lung and serum, in BMAL1-LysM-/- mice. In vitro studies demonstrated increased proinflammatory chemokine and mannose receptor expression in IL-4- as well as LPS-treated macrophages from BMAL1-LysM-/- mice compared with wild-type controls. This suggests that Bmal1 is a potent negative regulator in myeloid cells in the context of allergic asthma. Our findings might explain the increase in asthma incidents during the night, when BMAL1 expression is low.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Asma/complicaciones , Asma/metabolismo , Ritmo Circadiano , Hipersensibilidad/complicaciones , Hipersensibilidad/metabolismo , Células Mieloides/metabolismo , Animales , Asma/patología , Biomarcadores/metabolismo , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Eosinófilos/patología , Hipersensibilidad/patología , Mediadores de Inflamación/metabolismo , Interleucina-5/metabolismo , Pulmón/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Neumonía/complicaciones , Neumonía/metabolismo , Neumonía/patología
14.
Immunology ; 149(2): 225-37, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27353657

RESUMEN

Macrophage colony-stimulating factor 1 (CSF-1) plays a critical role in the differentiation of mononuclear phagocytes from bone marrow precursors, and maturing monocytes and macrophages exhibit increased expression of the CSF-1 receptor, CSF1R. The expression of CSF1R is tightly regulated by transcription factors and epigenetic mechanisms. We previously showed that prostaglandin E2 and subsequent activation of protein kinase A (PKA) inhibited CSF1R expression and macrophage maturation. Here, we examine the DNA methylation changes that occur at the Csf1r locus during macrophage maturation in the presence or absence of activated PKA. Murine bone marrow cells were matured to macrophages by incubating cells with CSF-1-containing conditioned medium for up to 6 days in the presence or absence of the PKA agonist 6-bnz-cAMP. DNA methylation of Csf1r promoter and enhancer regions was assayed by bisulphite pyrosequencing. DNA methylation of Csf1r decreased during normal macrophage maturation in concert with an increase in Csf1r mRNA expression. Treatment with the PKA agonist inhibited Csf1r mRNA and protein expression, and increased DNA methylation at the Csf1r promoter. This was associated with decreased binding of the transcription factor PU.1 to the Csf1r promoter. Treatment with the PKA agonist inhibited the responsiveness of macrophages to CSF-1. Levels of endogenous PKA activity decreased during normal macrophage maturation, suggesting that attenuation of this signalling pathway contributes to the increase in CSF1R expression during macrophage maturation. Together, these results demonstrate that macrophage maturation is accompanied by Csf1r hypomethylation, and illustrates for the first time the ability of PKA to increase Csf1r DNA methylation.


Asunto(s)
Células de la Médula Ósea/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Metilación de ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Transducción de Señal/genética , Transactivadores/genética , Transactivadores/metabolismo
15.
J Immunol ; 193(8): 4245-53, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25225663

RESUMEN

The role and origin of alveolar macrophages (AMs) in asthma are incompletely defined. We sought to clarify these issues in the context of acute allergic lung inflammation using house dust mite and OVA murine models. Use of liposomal clodronate to deplete resident AMs (rAMs) resulted in increased levels of inflammatory cytokines and eosinophil numbers in lavage fluid and augmented the histopathologic evidence of lung inflammation, suggesting a suppressive role for rAMs. Lung digests of asthmatic mice revealed an increased percentage of Ly6C(high)/CD11b(pos) inflammatory monocytes. Clodronate depletion of circulating monocytes, by contrast, resulted in an attenuation of allergic inflammation. A CD45.1/CD45.2 chimera model demonstrated that recruitment at least partially contributes to the AM pool in irradiated nonasthmatic mice, but its contribution was no greater in asthma. Ki-67 staining of AMs supported a role for local proliferation, which was increased in asthma. Our data demonstrate that rAMs dampen, whereas circulating monocytes promote, early events in allergic lung inflammation. Moreover, maintenance of the AM pool in the early stages of asthmatic inflammation depends on local proliferation, but not recruitment.


Asunto(s)
Asma/inmunología , Inflamación/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Alérgenos/inmunología , Alveolitis Alérgica Extrínseca/inmunología , Animales , Antígenos Ly/biosíntesis , Líquido del Lavado Bronquioalveolar/citología , Antígeno CD11b/biosíntesis , Proliferación Celular , Ácido Clodrónico/farmacología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Antígenos Comunes de Leucocito/genética , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Neumonía/inmunología , Pyroglyphidae/inmunología
16.
FASEB J ; 28(1): 56-66, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24072780

RESUMEN

Prostaglandin E2 (PGE2) regulates numerous biological processes by modulating transcriptional activation, epigenetic control, proteolysis, and secretion of various proteins. Scar formation depends on fibroblast elaboration of matrix proteins such as collagen, and this process is strongly suppressed by PGE2 through activation of cAMP-dependent protein kinase A (PKA). However, the actual mechanism by which PGE2-PKA signaling inhibits collagen expression in fibroblasts has never been delineated, and that was the objective of this study. PGE2 unexpectedly induced a rapid reduction in procollagen I protein expression in adult lung fibroblasts, with a half-maximum effect at 1.5 h. This effect reflected its inhibition of translation rather than transcription. Global protein synthesis was also inhibited by PGE2. This action was mediated by PKA and involved both activation of ribosomal protein (rpS6) and suppression of mammalian target of rapamycin (mTOR). Similar effects of PGE2 were demonstrated in mouse peritoneal macrophages (PMs). These findings identify inhibition of translation as a new mechanism by which PGE2 regulates cellular function and a novel example of translational inhibition mediated by opposing actions on two distinct translational control pathways. Translational inhibition would be expected to contribute to dynamic alterations in cell function that accompany the changing PGE2 levels observed in disease states and with various pharmacotherapies.


Asunto(s)
Dinoprostona/metabolismo , Fibroblastos/metabolismo , Animales , Línea Celular , Colágeno Tipo I/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Pulmón/citología , Pulmón/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Transporte de Proteínas/fisiología , Interferencia de ARN , Proteína S6 Ribosómica/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
17.
J Allergy Clin Immunol ; 133(2): 379-87, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24075232

RESUMEN

BACKGROUND: Endogenous prostanoids have been suggested to modulate sensitization during experimental allergic asthma, but the specific role of prostaglandin (PG) E2 or of specific E prostanoid (EP) receptors is not known. OBJECTIVE: Here we tested the role of EP2 signaling in allergic asthma. METHODS: Wild-type (WT) and EP2(-/-) mice were subjected to ovalbumin sensitization and acute airway challenge. The PGE2 analog misoprostol was administered during sensitization in both genotypes. In vitro culture of splenocytes and flow-sorted dendritic cells and T cells defined the mechanism by which EP2 exerted its protective effect. Adoptive transfer of WT and EP2(-/-) CD4 T cells was used to validate the importance of EP2 expression on T cells. RESULTS: Compared with WT mice, EP2(-/-) mice had exaggerated airway inflammation in this model. Splenocytes and lung lymph node cells from sensitized EP2(-/-) mice produced more IL-13 than did WT cells, suggesting increased sensitization. In WT but not EP2(-/-) mice, subcutaneous administration of misoprostol during sensitization inhibited allergic inflammation. PGE2 decreased cytokine production and inhibited signal transducer and activator of transcription 6 phosphorylation by CD3/CD28-stimulated CD4(+) T cells. Coculture of flow cytometry-sorted splenic CD4(+) T cells and CD11c(+) dendritic cells from WT or EP2(-/-) mice suggested that the increased IL-13 production in EP2(-/-) mice was due to the lack of EP2 specifically on T cells. Adoptive transfer of CD4(+) EP2(-/-) T cells caused greater cytokine production in the lungs of WT mice than did transfer of WT CD4(+) T cells. CONCLUSION: We conclude that the PGE2-EP2 axis is an important endogenous brake on allergic airway inflammation and primarily targets T cells and that its agonism represents a potential novel therapeutic approach to asthma.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Dinoprostona/inmunología , Neumonía/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/inmunología , Traslado Adoptivo , Alérgenos , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Citocinas/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Noqueados , Misoprostol/farmacología , Ovalbúmina , Subtipo EP2 de Receptores de Prostaglandina E/genética , Bazo/inmunología
18.
Am J Respir Cell Mol Biol ; 51(2): 242-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24601788

RESUMEN

Alveolar macrophages (AMs) represent the first line of innate immune defense in the lung. AMs use pattern recognition receptors (PRRs) to sense pathogens. The best studied PRR is Toll-like receptor (TLR)4, which detects LPS from gram-negative bacteria. The lipid mediator prostaglandin (PG)E2 dampens AM immune responses by inhibiting the signaling events downstream of PRRs. We examined the effect of PGE2 on TLR4 expression in rat AMs. Although PGE2 did not reduce the mRNA levels of TLR4, it decreased TLR4 protein levels. The translation inhibitor cycloheximide reduced TLR4 protein levels with similar kinetics as PGE2, and its effects were not additive with those of the prostanoid, suggesting that PGE2 inhibits TLR at the translational level. The action of PGE2 could be mimicked by the direct stimulator of cAMP formation, forskolin, and involved E prostanoid receptor 2 ligation and cAMP-dependent activation of unanchored type I protein kinase A. Cells pretreated with PGE2 for 24 hours exhibited decreased TNF-α mRNA and protein levels in response to LPS stimulation. Knockdown of TLR4 protein by small interfering RNA to the levels achieved by PGE2 treatment likewise decreased TNF-α mRNA and protein in response to LPS, establishing the functional significance of this PGE2 effect. We provide the first evidence of a lipid mediator acting through its cognate G protein-coupled receptor to affect PRR translation. Because PGE2 is produced in abundance at sites of infection, its inhibitory effects on AM TLR4 expression have important implications for host defense in the lung.


Asunto(s)
Dinoprostona/metabolismo , Macrófagos Alveolares/metabolismo , Receptor Toll-Like 4/metabolismo , Transcripción Genética , Adenilil Ciclasas/metabolismo , Animales , AMP Cíclico/metabolismo , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/inmunología , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 4/genética , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Células U937
19.
Blood ; 119(10): 2358-67, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22234697

RESUMEN

Prostaglandin E(2) (PGE(2)) is a lipid mediator that acts by ligating 4 distinct G protein-coupled receptors, E prostanoid (EP) 1 to 4. Previous studies identified the importance of PGE(2) in regulating macrophage functions, but little is known about its effect on macrophage maturation. Macrophage maturation was studied in vitro in bone marrow cell cultures, and in vivo in a model of peritonitis. EP2 was the most abundant PGE(2) receptor expressed by bone marrow cells, and its expression further increased during macrophage maturation. EP2-deficient (EP2(-/-)) macrophages exhibited enhanced in vitro maturation compared with wild-type cells, as evidenced by higher F4/80 expression. An EP2 antagonist also increased maturation. In the peritonitis model, EP2(-/-) mice exhibited a higher percentage of F4/80(high)/CD11b(high) cells and greater expression of macrophage colony-stimulating factor receptor (M-CSFR) in both the blood and the peritoneal cavity. Subcutaneous injection of the PGE(2) analog misoprostol decreased M-CSFR expression in bone marrow cells and reduced the number of peritoneal macrophages in wild-type mice but not EP2(-/-) mice. The suppressive effect of EP2 ligation on in vitro macrophage maturation was mimicked by a selective protein kinase A agonist. Our findings reveal a novel role for PGE(2)/EP2/protein kinase A signaling in the suppression of macrophage maturation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Macrófagos/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Células Cultivadas , Femenino , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Misoprostol/farmacología , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/metabolismo , Antagonistas de Prostaglandina/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tioglicolatos , Xantonas/farmacología
20.
Front Immunol ; 15: 1328781, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550597

RESUMEN

Metabolic changes are coupled with alteration in protein glycosylation. In this review, we will focus on macrophages that are pivotal in the pathogenesis of pulmonary fibrosis and sarcoidosis and thanks to their adaptable metabolism are an attractive therapeutic target. Examples presented in this review demonstrate that protein glycosylation regulates metabolism-driven immune responses in macrophages, with implications for fibrotic processes and granuloma formation. Targeting proteins that regulate glycosylation, such as fucosyltransferases, neuraminidase 1 and chitinase 1 could effectively block immunometabolic changes driving inflammation and fibrosis, providing novel avenues for therapeutic interventions.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Fibrosis Pulmonar , Sarcoidosis , Humanos , Glicosilación , Enfermedades Pulmonares Intersticiales/metabolismo , Fibrosis Pulmonar/etiología , Sarcoidosis/metabolismo , Fibrosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA