Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970705

RESUMEN

Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRß, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRß as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.


Asunto(s)
Homeostasis , Receptores X del Hígado , Macrófagos Alveolares , Neumonía , Surfactantes Pulmonares , Transducción de Señal , Animales , Receptores X del Hígado/metabolismo , Receptores X del Hígado/genética , Surfactantes Pulmonares/metabolismo , Ratones , Neumonía/metabolismo , Neumonía/patología , Macrófagos Alveolares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Pulmón/metabolismo , Pulmón/patología , Células Epiteliales Alveolares/metabolismo , Asma/metabolismo , Asma/patología , Asma/genética , Colesterol/metabolismo , Metabolismo de los Lípidos , Fagocitosis
2.
Nucleic Acids Res ; 50(22): 13063-13082, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36464162

RESUMEN

The glucocorticoid receptor (GR) is a ubiquitously expressed transcription factor that controls metabolic and homeostatic processes essential for life. Although numerous crystal structures of the GR ligand-binding domain (GR-LBD) have been reported, the functional oligomeric state of the full-length receptor, which is essential for its transcriptional activity, remains disputed. Here we present five new crystal structures of agonist-bound GR-LBD, along with a thorough analysis of previous structural work. We identify four distinct homodimerization interfaces on the GR-LBD surface, which can associate into 20 topologically different homodimers. Biologically relevant homodimers were identified by studying a battery of GR point mutants including crosslinking assays in solution, quantitative fluorescence microscopy in living cells, and transcriptomic analyses. Our results highlight the relevance of non-canonical dimerization modes for GR, especially of contacts made by loop L1-3 residues such as Tyr545. Our work illustrates the unique flexibility of GR's LBD and suggests different dimeric conformations within cells. In addition, we unveil pathophysiologically relevant quaternary assemblies of the receptor with important implications for glucocorticoid action and drug design.


Asunto(s)
Glucocorticoides , Receptores de Glucocorticoides , Receptores de Glucocorticoides/metabolismo , Ligandos , Unión Proteica , Dimerización
3.
Semin Cancer Biol ; 73: 58-75, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33309851

RESUMEN

Nuclear receptors (NRs) are a superfamily of ligand-activated transcription factors that act as biological sensors and use a combination of mechanisms to modulate positively and negatively gene expression in a spatial and temporal manner. The highly orchestrated biological actions of several NRs influence the proliferation, differentiation, and apoptosis of many different cell types. Synthetic ligands for several NRs have been the focus of extensive drug discovery efforts for cancer intervention. This review summarizes the roles in tumour growth and metastasis of several relevant NR family members, namely androgen receptor (AR), estrogen receptor (ER), glucocorticoid receptor (GR), thyroid hormone receptor (TR), retinoic acid receptors (RARs), retinoid X receptors (RXRs), peroxisome proliferator-activated receptors (PPARs), and liver X receptors (LXRs). These studies are key to develop improved therapeutic agents based on novel modes of action with reduced side effects and overcoming resistance.


Asunto(s)
Hormonas , Lípidos , Neoplasias , Receptores Citoplasmáticos y Nucleares , Animales , Humanos
4.
PLoS Genet ; 15(9): e1008399, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31527905

RESUMEN

Antimicrobial resistance (AMR) is currently one of the most important challenges to the treatment of bacterial infections. A critical issue to combat AMR is to restrict its spread. In several instances, bacterial plasmids are involved in the global spread of AMR. Plasmids belonging to the incompatibility group (Inc)HI are widespread in Enterobacteriaceae and most of them express multiple antibiotic resistance determinants. They play a relevant role in the recent spread of colistin resistance. We present in this report novel findings regarding IncHI plasmid conjugation. Conjugative transfer in liquid medium of an IncHI plasmid requires expression of a plasmid-encoded, large-molecular-mass protein that contains an Ig-like domain. The protein, termed RSP, is encoded by a gene (ORF R0009) that maps in the Tra2 region of the IncHI1 R27 plasmid. The RSP protein is exported outside the cell by using the plasmid-encoded type IV secretion system that is also used for its transmission to new cells. Expression of the protein reduces cell motility and enables plasmid conjugation. Flagella are one of the cellular targets of the RSP protein. The RSP protein is required for a high rate of plasmid transfer in both flagellated and nonflagellated Salmonella cells. This effect suggests that RSP interacts with other cellular structures as well as with flagella. These unidentified interactions must facilitate mating pair formation and, hence, facilitate IncHI plasmid conjugation. Due to its location on the outer surfaces of the bacterial cell, targeting the RSP protein could be a means of controlling IncHI plasmid conjugation in natural environments or of combatting infections caused by AMR enterobacteria that harbor IncHI plasmids.


Asunto(s)
Conjugación Genética/genética , Dominios de Inmunoglobulinas/genética , Factores R/genética , Secuencia de Aminoácidos , Antibacterianos/farmacología , Bacterias/genética , Proteínas Bacterianas/genética , Farmacorresistencia Microbiana/genética , Dominios de Inmunoglobulinas/fisiología , Plásmidos/genética , Salmonella/genética
5.
Cancer Immunol Immunother ; 68(4): 673-685, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30569204

RESUMEN

Many pathogens, ranging from viruses to multicellular parasites, promote expansion of MDSCs, which are myeloid cells that exhibit immunosuppressive features. The roles of MDSCs in infection depend on the class and virulence mechanisms of the pathogen, the stage of the disease, and the pathology associated with the infection. This work compiles evidence supported by functional assays on the roles of different subsets of MDSCs in acute and chronic infections, including pathogen-associated malignancies, and discusses strategies to modulate MDSC dynamics to benefit the host.


Asunto(s)
Enfermedades Transmisibles/etiología , Enfermedades Transmisibles/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Enfermedad Aguda , Animales , Biomarcadores , Enfermedad Crónica , Enfermedades Transmisibles/tratamiento farmacológico , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunomodulación , Terapia Molecular Dirigida , Células Supresoras de Origen Mieloide/efectos de los fármacos
6.
J Immunol ; 190(12): 6520-32, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686490

RESUMEN

Liver X receptors (LXRs) exert key functions in lipid homeostasis and in control of inflammation. In this study we have explored the impact of LXR activation on the macrophage response to the endogenous inflammatory cytokine IFN-γ. Transcriptional profiling studies demonstrate that ∼38% of the IFN-γ-induced transcriptional response is repressed by LXR activation in macrophages. LXRs also mediated inhibitory effects on selected IFN-γ-induced genes in primary microglia and in a model of IFN-γ-induced neuroinflammation in vivo. LXR activation resulted in reduced STAT1 recruitment to the promoters tested in this study without affecting STAT1 phosphorylation. A closer look into the mechanism revealed that SUMOylation of LXRs, but not the presence of nuclear receptor corepressor 1, was required for repression of the NO synthase 2 promoter. We have also analyzed whether IFN-γ signaling exerts reciprocal effects on LXR targets. Treatment with IFN-γ inhibited, in a STAT1-dependent manner, the LXR-dependent upregulation of selective targets, including ATP-binding cassette A1 (ABCA1) and sterol response element binding protein 1c. Downregulation of ABCA1 expression correlated with decreased cholesterol efflux to apolipoprotein A1 in macrophages stimulated with IFN-γ. The inhibitory effects of IFN-γ on LXR signaling did not involve reduced binding of LXR/retinoid X receptor heterodimers to target gene promoters. However, overexpression of the coactivator CREB-binding protein/p300 reduced the inhibitory actions of IFN-γ on the Abca1 promoter, suggesting that competition for CREB-binding protein may contribute to STAT1-dependent downregulation of LXR targets. The results from this study suggest an important level of bidirectional negative cross-talk between IFN-γ/STAT1 and LXRs with implications both in the control of IFN-γ-mediated immune responses and in the regulation of lipid metabolism.


Asunto(s)
Interferón gamma/inmunología , Macrófagos/inmunología , Receptores Nucleares Huérfanos/inmunología , Receptor Cross-Talk/inmunología , Factor de Transcripción STAT1/inmunología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/inmunología , Inflamación/inmunología , Metabolismo de los Lípidos/fisiología , Receptores X del Hígado , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Nucleares Huérfanos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Transcriptoma
7.
Circ Res ; 111(11): 1459-69, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22931956

RESUMEN

RATIONALE: Psychological stress is associated with an increased risk of cardiovascular diseases. However, the connecting mechanisms of the stress-inducing activation of the hypothalamic-pituitary-adrenal axis with atherosclerosis are not well-understood. OBJECTIVE: To study the effect of acute psychological stress on reverse cholesterol transport (RCT), which transfers peripheral cholesterol to the liver for its ultimate fecal excretion. METHODS AND RESULTS: C57Bl/6J mice were exposed to restraint stress for 3 hours to induce acute psychological stress. RCT in vivo was quantified by measuring the transfer of [(3)H]cholesterol from intraperitoneally injected mouse macrophages to the lumen of the small intestine within the stress period. Surprisingly, stress markedly increased the contents of macrophage-derived [(3)H]cholesterol in the intestinal lumen. In the stressed mice, intestinal absorption of [(14)C]cholesterol was significantly impaired, the intestinal mRNA expression level of peroxisome proliferator-activated receptor-α increased, and that of the sterol influx transporter Niemann-Pick C1-like 1 decreased. The stress-dependent effects on RCT rate and peroxisome proliferator-activated receptor-α gene expression were fully mimicked by administration of the stress hormone corticosterone (CORT) to nonstressed mice, and they were blocked by the inhibition of CORT synthesis in stressed mice. Moreover, the intestinal expression of Niemann-Pick C1-like 1 protein decreased when circulating levels of CORT increased. Of note, when either peroxisome proliferator-activated receptor α or liver X receptor α knockout mice were exposed to stress, the RCT rate remained unchanged, although plasma CORT increased. This indicates that activities of both transcription factors were required for the RCT-accelerating effect of stress. CONCLUSIONS: Acute psychological stress accelerated RCT by compromising intestinal cholesterol absorption. The present results uncover a novel functional connection between the hypothalamic-pituitary-adrenal axis and RCT that can be triggered by a stress-induced increase in circulating CORT.


Asunto(s)
Colesterol/metabolismo , Corticosterona/sangre , Estrés Psicológico/fisiopatología , Enfermedad Aguda , Animales , Transporte Biológico/efectos de los fármacos , Western Blotting , Línea Celular , Colesterol/farmacocinética , Corticosterona/farmacología , Femenino , Expresión Génica , Humanos , Absorción Intestinal/fisiología , Intestino Delgado/metabolismo , Lípidos/sangre , Hígado/metabolismo , Receptores X del Hígado , Macrófagos/metabolismo , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Immunol ; 186(8): 4656-67, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21398609

RESUMEN

Macrophages serve essential functions as regulators of immunity and homeostasis, and their proliferation contributes to pathogenesis of certain disorders. In this report, we show that induction of macrophage proliferation by the growth factor M-CSF is negatively modulated by agonists that activate the nuclear receptor liver X receptor (LXR), both in vitro and in vivo. Both isoforms LXR α and ß are involved in the antiproliferative actions of LXR ligands in macrophages. In contrast, M-CSF does not exert negative effects on LXR-mediated gene expression. Treatment with LXR agonists results in the accumulation of macrophages in the G(0)/G(1) phase of the cell cycle without affecting ERK-1/2 phosphorylation. The use of small interfering RNA or genetically modified mice revealed that, in contrast to other cellular models, functional expression of either the cyclin-dependent kinase inhibitor p27KIP1 or the cholesterol transporters ATP-binding cassette A1 or ATP-binding cassette G1 was not required for the antiproliferative effects of LXR agonists in macrophages. Western blot analysis revealed that protein expression of key molecules that regulate progression through the cell cycle, such as cyclins D1 and B1 and cyclin-dependent kinases 2 and 4, was downregulated upon LXR activation. These observations suggest a role for LXR agonists in limiting macrophage proliferative responses associated to pathogenic disorders.


Asunto(s)
Proliferación Celular , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Western Blotting , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Ciclina B1/metabolismo , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Células HEK293 , Humanos , Hidrocarburos Fluorados/farmacología , Células L , Receptores X del Hígado , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/genética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sulfonamidas/farmacología
9.
EBioMedicine ; 91: 104555, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37054630

RESUMEN

BACKGROUND: Reprogramming of immunosuppressive tumor-associated macrophages (TAMs) presents an attractive therapeutic strategy in cancer. The aim of this study was to explore the role of macrophage CD5L protein in TAM activity and assess its potential as a therapeutic target. METHODS: Monoclonal antibodies (mAbs) against recombinant CD5L were raised by subcutaneous immunization of BALB/c mice. Peripheral blood monocytes were isolated from healthy donors and stimulated with IFN/LPS, IL4, IL10, and conditioned medium (CM) from different cancer cell lines in the presence of anti-CD5L mAb or controls. Subsequently, phenotypic markers, including CD5L, were quantified by flow cytometry, IF and RT-qPCR. Macrophage CD5L protein expression was studied in 55 human papillary lung adenocarcinoma (PAC) samples by IHC and IF. Anti-CD5L mAb and isotype control were administered intraperitoneally into a syngeneic Lewis Lung Carcinoma mouse model and tumor growth was measured. Tumor microenvironment (TME) changes were determined by flow cytometry, IHC, IF, Luminex, RNAseq and RT-qPCR. FINDINGS: Cancer cell lines CM induced an immunosuppressive phenotype (increase in CD163, CD206, MERTK, VEGF and CD5L) in cultured macrophages. Accordingly, high TAM expression of CD5L in PAC was associated with poor patient outcome (Log-rank (Mantel-Cox) test p = 0.02). We raised a new anti-CD5L mAb that blocked the immunosuppressive phenotype of macrophages in vitro. Its administration in vivo inhibited tumor progression of lung cancer by altering the intratumoral myeloid cell population profile and CD4+ T-cell exhaustion phenotype, thereby significantly modifying the TME and increasing the inflammatory milieu. INTERPRETATION: CD5L protein plays a key function in modulating the activity of macrophages and their interactions within the TME, which supports its role as a therapeutic target in cancer immunotherapy. FUNDING: For a full list of funding bodies, please see the Acknowledgements.


Asunto(s)
Neoplasias Pulmonares , Macrófagos , Animales , Humanos , Ratones , Línea Celular Tumoral , Inmunoterapia , Neoplasias Pulmonares/terapia , Macrófagos/metabolismo , Monocitos , Células Mieloides/patología , Microambiente Tumoral
10.
Cancer Res ; 81(4): 968-985, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33361391

RESUMEN

Liver X receptors (LXR) are transcription factors from the nuclear receptor family that are activated by oxysterols and synthetic high-affinity agonists. In this study, we assessed the antitumor effects of synthetic LXR agonist TO901317 in a murine model of syngeneic Lewis Lung carcinoma. Treatment with TO901317 inhibited tumor growth in wild-type, but not in LXR-deficient mice, indicating that the antitumor effects of the agonist depends on functional LXR activity in host cells. Pharmacologic activation of the LXR pathway reduced the intratumoral abundance of regulatory T cells (Treg) and the expression of the Treg-attracting chemokine Ccl17 by MHCIIhigh tumor-associated macrophages (TAM). Moreover, gene expression profiling indicated a broad negative impact of the LXR agonist on other mechanisms used by TAM for the maintenance of an immunosuppressive environment. In studies exploring the macrophage response to GM-CSF or IL4, activated LXR repressed IRF4 expression, resulting in subsequent downregulation of IRF4-dependent genes including Ccl17. Taken together, this work reveals the combined actions of the LXR pathway in the control of TAM responses that contribute to the antitumoral effects of pharmacologic LXR activation. Moreover, these data provide new insights for the development of novel therapeutic options for the treatment of cancer. SIGNIFICANCE: This study reveals unrecognized roles of LXR in the transcriptional control of the tumor microenvironment and suggests use of a synthetic LXR agonist as a novel therapeutic strategy to stimulate antitumor activity.


Asunto(s)
Benzoatos/farmacología , Bencilaminas/farmacología , Hidrocarburos Fluorados/farmacología , Sulfonamidas/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/efectos de los fármacos , Animales , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Receptores X del Hígado/agonistas , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Células RAW 264.7 , Linfocitos T Reguladores/patología , Transcriptoma/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/patología
11.
Eur J Immunol ; 39(2): 514-26, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19130475

RESUMEN

Macrophages are recruited from the blood stream to the inflammatory loci to carry out their functional activities. In an early phase of the cell cycle, macrophages become activated by Th1-type cytokines (i.e. IFN-gamma), thereby producing several factors (cytokines, NO, etc.) and developing pro-inflammatory activities. When bacteria and apoptotic bodies are removed, through the interaction with Th2-type cytokines (i.e. IL-4), macrophages become anti-inflammatory and repair damaged tissues. Incubation of bone-marrow-derived macrophages with IFN-gamma or IL-4 blocked their proliferation. While M-CSF withdrawal caused cell cycle arrest at the early G(1) phase, treatment of macrophages with IFN-gamma or IL-4 caused this arrest later, at the G(1)/S boundary. Proliferation arrest was not due to an induction of apoptosis. IFN-gamma and IL-4 induced the expression of the cyclin-dependent kinase (Cdk) inhibitor p21(Waf1). Using KO mice and iRNA experiments, we found that p21(Waf1)is required for IL-4- but not for IFN-gamma-dependent inhibition of macrophage proliferation. IL-4 inhibited M-CSF-dependent Cdk-2 and Cdk-4 activities, which are necessary for entry and passage through the S phase of the cell cycle. The signal transduction used to induce the expression of p21(Waf1)after interaction of IL-4 with the corresponding receptor was mediated by STAT6. Thus, IL-4 and IFN-gamma blocked M-CSF-induced macrophage proliferation through distinct mechanisms.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Interleucina-4/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Factor de Transcripción STAT6/metabolismo , Animales , Antivirales/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/inmunología , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Interferón gamma/inmunología , Interferón gamma/farmacología , Interleucina-4/farmacología , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
12.
Blood ; 112(8): 3274-82, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18682602

RESUMEN

Macrophages have the capacity to proliferate in response to specific growth factors, such as macrophage-colony stimulating factor (M-CSF). In the presence of several cytokines and activating factors, macrophages undergo growth arrest, become activated, and participate in the development of an immune response. We have previously observed that activation of extracellularly regulated kinase 1/2 (ERK-1/2) is required for macrophage proliferation in response to growth factors. A short and early pattern of ERK activity correlated with the proliferative response. In contrast, slightly prolonged patterns of activity of these kinases were induced by signals that lead to macrophage activation and growth arrest. IFN-gamma is the main endogenous Th1-type macrophage activator. Here we report that stimulation with IFN-gamma prolongs the pattern of ERK activity induced by M-CSF in macrophages. These effects correlate with IFN-gamma-mediated inhibition of the expression of several members of the MAPK phosphatase family, namely MKP-1, -2, and -4. Moreover, inhibition of MKP-1 expression using siRNA technology or synthetic inhibitors also led to elongated ERK activity and significant blockage of M-CSF-dependent proliferation. These data suggest that subtle changes in the time course of activity of members of the MAPK family contribute to the antiproliferative effects of IFN-gamma in macrophages.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/biosíntesis , Regulación Enzimológica de la Expresión Génica , Interferón gamma/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/enzimología , Animales , Células de la Médula Ósea/citología , Proteínas de Ciclo Celular , Proliferación Celular , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Fenotipo , Transducción de Señal
13.
Cells ; 9(1)2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31963337

RESUMEN

CD38 is a multifunctional protein widely expressed in cells from the immune system and as a soluble form in biological fluids. CD38 expression is up-regulated by an array of inflammatory mediators, and it is frequently used as a cell activation marker. Studies in animal models indicate that CD38 functional expression confers protection against infection by several bacterial and parasitic pathogens. In addition, infectious complications are associated with anti-CD38 immunotherapy. Although CD38 displays receptor and enzymatic activities that contribute to the establishment of an effective immune response, recent work raises the possibility that CD38 might also enhance the immunosuppressive potential of regulatory leukocytes. This review integrates the current knowledge on the diversity of functions mediated by CD38 in the host defense to infection.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Infecciones/inmunología , ADP-Ribosil Ciclasa 1/deficiencia , Animales , Susceptibilidad a Enfermedades , Humanos , Inmunoterapia , Infecciones/patología , Infecciones/terapia , Fagocitosis , Fenotipo
14.
Curr Opin Pharmacol ; 53: 55-65, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32599447

RESUMEN

Liver X receptors (LXRs) are transcription factors from the nuclear receptor family that can be pharmacologically activated by high-affinity agonists. LXR activation exerts a combination of metabolic and anti-inflammatory actions that result in the modulation of immune responses and in the amelioration of inflammatory disorders. In addition, LXR agonists modulate the metabolism of infected cells and limit the infectivity and/or growth of several pathogens. This review gives an overview of the recent advances in understanding the complexity of the mechanisms through which the LXR pathway controls inflammation and host-cell pathogen interaction.


Asunto(s)
Receptores X del Hígado/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Infecciones/inmunología , Inflamación/inmunología
15.
Antioxidants (Basel) ; 9(11)2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-33233455

RESUMEN

The potential of nicotinamide (NAM) to prevent atherosclerosis has not yet been examined. This study investigated the effect of NAM supplementation on the development of atherosclerosis in a mouse model of the disease. The development of aortic atherosclerosis was significantly reduced (NAM low dose: 45%; NAM high dose: 55%) in NAM-treated, apolipoprotein (Apo)E-deficient mice challenged with a Western diet for 4 weeks. NAM administration significantly increased (1.8-fold) the plasma concentration of proatherogenic ApoB-containing lipoproteins in NAM high-dose (HD)-treated mice compared with untreated mice. However, isolated ApoB-containing lipoproteins from NAM HD mice were less prone to oxidation than those of untreated mice. This result was consistent with the decreased (1.5-fold) concentration of oxidized low-density lipoproteins in this group. Immunohistochemical staining of aortas from NAM-treated mice showed significantly increased levels of IL-10 (NAM low-dose (LD): 1.3-fold; NAM HD: 1.2-fold), concomitant with a significant decrease in the relative expression of TNFα (NAM LD: -44%; NAM HD: -57%). An improved anti-inflammatory pattern was reproduced in macrophages cultured in the presence of NAM. Thus, dietary NAM supplementation in ApoE-deficient mice prevented the development of atherosclerosis and improved protection against ApoB-containing lipoprotein oxidation and aortic inflammation.

16.
Methods Mol Biol ; 1951: 135-141, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30825149

RESUMEN

Macrophages are phagocytic cells that actively engulf and kill microorganisms within a specialized phagolysosomal system. Several pathogenic bacteria, however, actively co-opt host mechanisms and escape from microbial digestion to establish intracellular replication within macrophages. This chapter highlights detailed protocols to measure the effects of the LXR pathway on bacterial infection of murine bone marrow-derived macrophages.


Asunto(s)
Infecciones Bacterianas/metabolismo , Receptores X del Hígado/agonistas , Receptores X del Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Animales , Infecciones Bacterianas/microbiología , Biomarcadores , Citometría de Flujo , Inmunofenotipificación , Ratones , Microscopía Confocal , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/metabolismo
17.
Sci Rep ; 8(1): 6535, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29695842

RESUMEN

Metal limitation is a common situation during infection and can have profound effects on the pathogen's success. In this report, we examine the role of zinc limitation in the expression of a virulence factor in uropathogenic Escherichia coli. The pyelonephritis isolate J96 carries two hlyCABD operons that encode the RTX toxin α-hemolysin. While the coding regions of both operons are largely conserved, the upstream sequences, including the promoters, are unrelated. We show here that the two hlyCABD operons are differently regulated. The hly II operon is efficiently silenced in the presence of zinc and highly expressed when zinc is limited. In contrast, the hly I operon does not respond to zinc limitation. Genetic studies reveal that zinc-responsive regulation of the hly II operon is controlled by the Zur metalloregulatory protein. A Zur binding site was identified in the promoter sequence of the hly II operon, and we observe direct binding of Zur to this promoter region. Moreover, we find that Zur regulation of the hly II operon modulates the ability of E. coli J96 to induce a cytotoxic response in host cell lines in culture. Our report constitutes the first description of the involvement of the zinc-sensing protein Zur in directly modulating the expression of a virulence factor in bacteria.


Asunto(s)
Proteínas Hemolisinas/genética , Escherichia coli Uropatógena/genética , Factores de Virulencia/genética , Virulencia/genética , Zinc/metabolismo , Proteínas Bacterianas/genética , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Operón/genética , Regiones Promotoras Genéticas/genética , Pielonefritis/genética
18.
Mol Cell Biol ; 38(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29507185

RESUMEN

The liver X receptors (LXRs) are ligand-activated nuclear receptors with established roles in the maintenance of lipid homeostasis in multiple tissues. LXRs exert additional biological functions as negative regulators of inflammation, particularly in macrophages. However, the transcriptional responses controlled by LXRs in other myeloid cells, such as dendritic cells (DCs), are still poorly understood. Here we used gain- and loss-of-function models to characterize the impact of LXR deficiency on DC activation programs. Our results identified an LXR-dependent pathway that is important for DC chemotaxis. LXR-deficient mature DCs are defective in stimulus-induced migration in vitro and in vivo Mechanistically, we show that LXRs facilitate DC chemotactic signaling by regulating the expression of CD38, an ectoenzyme important for leukocyte trafficking. Pharmacological or genetic inactivation of CD38 activity abolished the LXR-dependent induction of DC chemotaxis. Using the low-density lipoprotein receptor-deficient (LDLR-/-) LDLR-/- mouse model of atherosclerosis, we also demonstrated that hematopoietic CD38 expression is important for the accumulation of lipid-laden myeloid cells in lesions, suggesting that CD38 is a key factor in leukocyte migration during atherogenesis. Collectively, our results demonstrate that LXRs are required for the efficient emigration of DCs in response to chemotactic signals during inflammation.


Asunto(s)
Quimiotaxis/fisiología , Células Dendríticas/fisiología , Receptores X del Hígado/fisiología , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Células Cultivadas , Células Dendríticas/citología , Inflamación , Metabolismo de los Lípidos , Receptores X del Hígado/genética , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares , Transducción de Señal
19.
J Clin Invest ; 114(11): 1564-76, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15578089

RESUMEN

PPARalpha, beta/delta, and gamma regulate genes involved in the control of lipid metabolism and inflammation and are expressed in all major cell types of atherosclerotic lesions. In vitro studies have suggested that PPARs exert antiatherogenic effects by inhibiting the expression of proinflammatory genes and enhancing cholesterol efflux via activation of the liver X receptor-ABCA1 (LXR-ABCA1) pathway. To investigate the potential importance of these activities in vivo, we performed a systematic analysis of the effects of PPARalpha, beta, and gamma agonists on foam-cell formation and atherosclerosis in male LDL receptor-deficient (LDLR(-/-)) mice. Like the PPARgamma agonist, a PPARalpha-specific agonist strongly inhibited atherosclerosis, whereas a PPARbeta-specific agonist failed to inhibit lesion formation. In concert with their effects on atherosclerosis, PPARalpha and PPARgamma agonists, but not the PPARbeta agonist, inhibited the formation of macrophage foam cells in the peritoneal cavity. Unexpectedly, PPARalpha and PPARgamma agonists inhibited foam-cell formation in vivo through distinct ABCA1-independent pathways. While inhibition of foam-cell formation by PPARalpha required LXRs, activation of PPARgamma reduced cholesterol esterification, induced expression of ABCG1, and stimulated HDL-dependent cholesterol efflux in an LXR-independent manner. In concert, these findings reveal receptor-specific mechanisms by which PPARs influence macrophage cholesterol homeostasis. In the future, these mechanisms may be exploited pharmacologically to inhibit the development of atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Células Espumosas/fisiología , PPAR alfa/metabolismo , PPAR delta/metabolismo , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Aorta/patología , Arteriosclerosis/patología , Colesterol/metabolismo , Colesterol en la Dieta , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Humanos , Receptores X del Hígado , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptores Nucleares Huérfanos , PPAR alfa/agonistas , PPAR alfa/genética , PPAR delta/agonistas , PPAR delta/genética , PPAR gamma/agonistas , PPAR gamma/genética , PPAR-beta/agonistas , PPAR-beta/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Triglicéridos/metabolismo
20.
Mol Cell Biol ; 23(16): 5780-9, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12897148

RESUMEN

Liver X receptors (LXRs) regulate the expression of genes involved in cholesterol and fatty acid homeostasis, including the genes for ATP-binding cassette transporter A1 (ABCA1) and sterol response element binding protein 1 (SREBP1). Loss of LXR leads to derepression of the ABCA1 gene in macrophages and the intestine, while the SREBP1c gene remains transcriptionally silent. Here we report that high-density-lipoprotein (HDL) cholesterol levels are increased in LXR-deficient mice, suggesting that derepression of ABCA1 and possibly other LXR target genes in selected tissues is sufficient to result in enhanced HDL biogenesis at the whole-body level. We provide several independent lines of evidence indicating that the repressive actions of LXRs are dependent on interactions with the nuclear receptor corepressor (NCoR) and the silencing mediator of retinoic acid and thyroid hormone receptors (SMRT). While dissociation of NCoR and SMRT results in derepression of the ABCA1 gene in macrophages, it is not sufficient for derepression of the SREBP1c gene. These findings reveal differential requirements for corepressors in the regulation of genes involved in cholesterol and fatty acid homeostasis and raise the possibility that these interactions may be exploited to develop synthetic ligands that selectively modulate LXR actions in vivo.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Northern Blotting , Western Blotting , Células de la Médula Ósea/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Diferenciación Celular , Línea Celular , Colesterol/metabolismo , HDL-Colesterol/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/biosíntesis , Silenciador del Gen , Genotipo , Ligandos , Receptores X del Hígado , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Nucleares , Co-Represor 1 de Receptor Nuclear , Receptores Nucleares Huérfanos , Pruebas de Precipitina , ARN/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Hormonas Tiroideas/metabolismo , Transcripción Genética , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA