Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 384(6691): eabo7027, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38574142

RESUMEN

Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.


Asunto(s)
Apoptosis , Interleucina-4 , Macrófagos , Fagocitosis , Esquistosomiasis mansoni , Animales , Ratones , Apoptosis/inmunología , Hepatocitos/inmunología , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/inmunología , Ratones Noqueados , Neutrófilos/inmunología , Fagocitosis/inmunología , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/inmunología , Modelos Animales de Enfermedad
2.
Pharmacol Res ; 163: 105292, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33171305

RESUMEN

Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.


Asunto(s)
Angiotensina I/uso terapéutico , Antiinflamatorios/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Infecciones Neumocócicas/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Proteínas Proto-Oncogénicas/inmunología , Receptores Acoplados a Proteínas G/inmunología , Células A549 , Angiotensina I/farmacología , Animales , Antiinflamatorios/farmacología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/inmunología , Perros , Humanos , Virus de la Influenza A , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Fragmentos de Péptidos/farmacología , Peroxidasa/inmunología , Fagocitosis/efectos de los fármacos , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Neumonía Viral/inmunología , Neumonía Viral/patología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Streptococcus pneumoniae
3.
Cell ; 183(1): 94-109.e23, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937105

RESUMEN

Cardiomyocytes are subjected to the intense mechanical stress and metabolic demands of the beating heart. It is unclear whether these cells, which are long-lived and rarely renew, manage to preserve homeostasis on their own. While analyzing macrophages lodged within the healthy myocardium, we discovered that they actively took up material, including mitochondria, derived from cardiomyocytes. Cardiomyocytes ejected dysfunctional mitochondria and other cargo in dedicated membranous particles reminiscent of neural exophers, through a process driven by the cardiomyocyte's autophagy machinery that was enhanced during cardiac stress. Depletion of cardiac macrophages or deficiency in the phagocytic receptor Mertk resulted in defective elimination of mitochondria from the myocardial tissue, activation of the inflammasome, impaired autophagy, accumulation of anomalous mitochondria in cardiomyocytes, metabolic alterations, and ventricular dysfunction. Thus, we identify an immune-parenchymal pair in the murine heart that enables transfer of unfit material to preserve metabolic stability and organ function. VIDEO ABSTRACT.


Asunto(s)
Macrófagos/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Anciano , Animales , Apoptosis , Autofagia , Femenino , Corazón/fisiología , Homeostasis , Humanos , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/fisiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Fagocitosis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa c-Mer/metabolismo
4.
Curr Opin Pharmacol ; 53: 18-26, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32361182

RESUMEN

Macrophages are immune myeloid cells with an extreme ability to modulate their phenotype in response to insults and/or pathogens. The immunomodulatory capacity of macrophages is also patent during development as they adapt their phenotype to the host tissue environment establishing the heterogeneous populations of tissue-resident macrophages. An important mechanism of immunomodulation in macrophages occurs through the regulation of transcriptional activity. Numerous transcription factors are associated with macrophage plasticity, among them, several nuclear receptors. The nuclear receptors Liver X Receptors (LXRα and LXRß) have also revealed as active players during macrophage adaptations in diverse scenarios. This review will address the different mechanisms by which LXRs contribute to immunomodulation in macrophages by connecting lipid metabolism and immunity through transcriptional regulation.


Asunto(s)
Inmunomodulación , Metabolismo de los Lípidos , Receptores X del Hígado/inmunología , Macrófagos/inmunología , Animales , Diferenciación Celular , Humanos
5.
J Exp Med ; 215(11): 2778-2795, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30282719

RESUMEN

Immune protection relies on the capacity of neutrophils to infiltrate challenged tissues. Naive tissues, in contrast, are believed to remain free of these cells and protected from their toxic cargo. Here, we show that neutrophils are endowed with the capacity to infiltrate multiple tissues in the steady-state, a process that follows tissue-specific dynamics. By focusing in two particular tissues, the intestine and the lungs, we find that neutrophils infiltrating the intestine are engulfed by resident macrophages, resulting in repression of Il23 transcription, reduced G-CSF in plasma, and reinforced activity of distant bone marrow niches. In contrast, diurnal accumulation of neutrophils within the pulmonary vasculature influenced circadian transcription in the lungs. Neutrophil-influenced transcripts in this organ were associated with carcinogenesis and migration. Consistently, we found that neutrophils dictated the diurnal patterns of lung invasion by melanoma cells. Homeostatic infiltration of tissues unveils a facet of neutrophil biology that supports organ function, but can also instigate pathological states.


Asunto(s)
Neoplasias Pulmonares/inmunología , Pulmón/inmunología , Melanoma/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Animales , Femenino , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Macrófagos/inmunología , Macrófagos/patología , Masculino , Melanoma/genética , Melanoma/patología , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Invasividad Neoplásica/inmunología , Neutrófilos/patología , Transcripción Genética/inmunología
6.
Cell Immunol ; 330: 151-158, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29779612

RESUMEN

Macrophage heterogeneity in the spleen has been long documented, with four subsets populating the different splenic compartments. The diverse environments on the splenic compartments determine their varied phenotype and functions. In the white pulp, highly phagocytic macrophages contribute to the generation of the immune response. The marginal zone contains two populations of macrophages, which also contribute to the immune response. Their strategic position in the bloodstream and their unique phenotype confer them a crucial role in the defense against blood borne pathogens, placing them at the crossroad between innate and adaptive immune responses. Macrophages in the red pulp are classically linked to homeostatic and metabolic functions in erythrocyte phagocytosis and iron recycling. We review here recent advances demonstrating the importance of macrophage ontogeny and organ development in determining the phenotype, transcriptional profile and, ultimately, the functions of the populations of splenic macrophages.


Asunto(s)
Metabolismo Energético/inmunología , Homeostasis/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Bazo/inmunología , Inmunidad Adaptativa/inmunología , Animales , Eritrocitos/inmunología , Humanos , Inmunidad Innata/inmunología , Bazo/citología , Bazo/metabolismo
7.
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
8.
J Exp Med ; 214(5): 1281-1296, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28432199

RESUMEN

Tissue-resident macrophages display varying phenotypic and functional properties that are largely specified by their local environment. One of these functions, phagocytosis, mediates the natural disposal of billions of cells, but its mechanisms and consequences within living tissues are poorly defined. Using a parabiosis-based strategy, we identified and isolated macrophages from multiple tissues as they phagocytosed blood-borne cellular material. Phagocytosis was circadianally regulated and mediated by distinct repertoires of receptors, opsonins, and transcription factors in macrophages from each tissue. Although the tissue of residence defined the core signature of macrophages, phagocytosis imprinted a distinct antiinflammatory profile. Phagocytic macrophages expressed CD206, displayed blunted expression of Il1b, and supported tissue homeostasis. Thus, phagocytosis is a source of macrophage heterogeneity that acts together with tissue-derived factors to preserve homeostasis.


Asunto(s)
Macrófagos/fisiología , Fagocitosis/fisiología , Animales , Femenino , Interleucina-1beta/metabolismo , Lectinas Tipo C/metabolismo , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Opsoninas/fisiología , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/fisiología
9.
Int J Hematol ; 99(6): 685-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24634109

RESUMEN

Two cellular systems of paramount importance for mammalian physiology, the myeloid and the hematopoietic, have received a great deal of attention in the past decade. Myeloid leukocytes, classically involved in mediating innate immune responses, are now known to regulate other important aspects of the organism's physiology, from development to regulation of metabolic functions. In parallel, many diverse cellular and molecular components have been identified in the bone marrow (BM) that are required for the regulation and lifelong preservation of hematopoietic stem and progenitor cells (HSPC). Since the production of blood and immune elements by these multipotent cells responds to environmental signals, it is not entirely surprising that the hematopoietic niches in which HSPC are located can in turn be regulated by the immune system. We review here recent evidence demonstrating that two components of the innate immune system, macrophages and neutrophils, regulate the function of the hematopoietic niche in ways that may favor both the retention and the release of HSPC from the BM. We propose that the highly migratory nature of neutrophils, the presence of a network of tissue-resident macrophages in the BM and possibly in other tissues, and the superb capacity of these innate immune cells to respond to stress endow them with regulatory functions that are ultimately relayed to the hematopoietic niche.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Inmunidad Innata , Nicho de Células Madre , Animales , Comunicación Celular , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo
10.
J Exp Med ; 210(11): 2321-36, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24081949

RESUMEN

Blood neutrophil homeostasis is essential for successful host defense against invading pathogens. Circulating neutrophil counts are positively regulated by CXCR2 signaling and negatively regulated by the CXCR4-CXCL12 axis. In particular, G-CSF, a known CXCR2 signaler, and plerixafor, a CXCR4 antagonist, have both been shown to correct neutropenia in human patients. G-CSF directly induces neutrophil mobilization from the bone marrow (BM) into the blood, but the mechanisms underlying plerixafor-induced neutrophilia remain poorly defined. Using a combination of intravital multiphoton microscopy, genetically modified mice and novel in vivo homing assays, we demonstrate that G-CSF and plerixafor work through distinct mechanisms. In contrast to G-CSF, CXCR4 inhibition via plerixafor does not result in neutrophil mobilization from the BM. Instead, plerixafor augments the frequency of circulating neutrophils through their release from the marginated pool present in the lung, while simultaneously preventing neutrophil return to the BM. Our study demonstrates for the first time that drastic changes in blood neutrophils can originate from alternative reservoirs other than the BM, while implicating a role for CXCR4-CXCL12 interactions in regulating lung neutrophil margination. Collectively, our data provides valuable insights into the fundamental regulation of neutrophil homeostasis, which may lead to the development of improved treatment regimens for neutropenic patients.


Asunto(s)
Médula Ósea/metabolismo , Movimiento Celular/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Pulmón/citología , Neutrófilos/citología , Receptores CXCR4/antagonistas & inhibidores , Animales , Bencilaminas , Médula Ósea/efectos de los fármacos , Ciclamas , Factor Estimulante de Colonias de Granulocitos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Recuento de Leucocitos , Macaca fascicularis , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica , Muramidasa/metabolismo , Músculo Esquelético/citología , Mutación/genética , Circulación Pulmonar , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores de Interleucina-8B/metabolismo
11.
Lymphat Res Biol ; 11(3): 187-92, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24044758

RESUMEN

BACKGROUND: Serious infections of the head and neck cause lymphedema that can lead to airway compromise and oropharyngeal obstruction. Lymphangiogenesis occurs in the head and neck during infection and after immunization. The goal of this project was to develop tools to image lymphatic vessels in living animals and to be able to isolate individual lymphatic endothelial cells in order to quantify changes in single cells caused by inflammation. METHODS: The ProxTom transgenic red-fluorescent reporter mouse was developed specifically for the purpose of imaging lymphatic vessels in vivo. Prox1 is a transcription factor that is necessary for lymphangiogenesis in development and for the maintenance of lymphatics in adulthood. Mice were immunized and their lymphatic vessels in lymph nodes were imaged in vivo. Individual lymphatic endothelial cells were isolated by means of their fluorescence. RESULTS: The ProxTom transgene has the red-fluorescent reporter td-Tomato under the control of Prox1 regulatory elements. tdTomato was faithfully expressed in lymphatic vessels coincident with endogenous Prox1 expression. We show lymphangiogenesis in vivo after immunization and demonstrate a method for the isolation of lymphatic endothelial cells by their tdTomato red-fluorescence. CONCLUSIONS: The faithful expression of the red-fluorescent reporter in the lymphatic vessels of ProxTom means that these mice have proven utility for in vivo study of lymphatic vessels in the immune response. ProxTom has been made available for distribution from the Jackson Laboratory: http://jaxmice.jax.org/strain/018128.html .


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/metabolismo , Linfangiogénesis , Vasos Linfáticos/metabolismo , Animales , Células Endoteliales/inmunología , Células Endoteliales/patología , Citometría de Flujo , Cabeza , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunización/métodos , Inflamación/genética , Inflamación/inmunología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Vasos Linfáticos/inmunología , Vasos Linfáticos/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Cuello , Oxazolona/inmunología , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína Fluorescente Roja
12.
Nat Immunol ; 14(8): 831-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23770640

RESUMEN

Macrophages are professional phagocytic cells that orchestrate innate immune responses and have considerable phenotypic diversity at different anatomical locations. However, the mechanisms that control the heterogeneity of tissue macrophages are not well characterized. Here we found that the nuclear receptor LXRα was essential for the differentiation of macrophages in the marginal zone (MZ) of the spleen. LXR-deficient mice were defective in the generation of MZ and metallophilic macrophages, which resulted in abnormal responses to blood-borne antigens. Myeloid-specific expression of LXRα or adoptive transfer of wild-type monocytes restored the MZ microenvironment in LXRα-deficient mice. Our results demonstrate that signaling via LXRα in myeloid cells is crucial for the generation of splenic MZ macrophages and identify an unprecedented role for a nuclear receptor in the generation of specialized macrophage subsets.


Asunto(s)
Hematopoyesis/inmunología , Macrófagos/inmunología , Receptores Nucleares Huérfanos/inmunología , Bazo/inmunología , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Diferenciación Celular/inmunología , Citometría de Flujo , Inmunidad Celular/inmunología , Inmunohistoquímica , Receptores X del Hígado , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Receptores Nucleares Huérfanos/agonistas , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos , Bazo/citología
13.
Cell ; 153(5): 1025-35, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706740

RESUMEN

Unique among leukocytes, neutrophils follow daily cycles of release from and migration back into the bone marrow, where they are eliminated. Because removal of dying cells generates homeostatic signals, we explored whether neutrophil elimination triggers circadian events in the steady state. Here, we report that the homeostatic clearance of neutrophils provides cues that modulate the physiology of the bone marrow. We identify a population of CD62L(LO) CXCR4(HI) neutrophils that have "aged" in the circulation and are eliminated at the end of the resting period in mice. Aged neutrophils infiltrate the bone marrow and promote reductions in the size and function of the hematopoietic niche. Modulation of the niche depends on macrophages and activation of cholesterol-sensing nuclear receptors and is essential for the rhythmic egress of hematopoietic progenitors into the circulation. Our results unveil a process that synchronizes immune and hematopoietic rhythms and expand the ascribed functions of neutrophils beyond inflammation. PAPERFLICK:


Asunto(s)
Médula Ósea/fisiología , Ritmo Circadiano , Neutrófilos/citología , Neutrófilos/fisiología , Animales , Movimiento Celular , Senescencia Celular , Femenino , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores Nucleares Huérfanos/metabolismo
14.
Biochim Biophys Acta ; 1812(8): 982-94, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21193033

RESUMEN

The liver X receptors (LXRα and LXRß) are members of the nuclear receptor family of transcription factors that play essential roles in the transcriptional control of lipid metabolism. LXRs are endogenously activated by modified forms of cholesterol known as oxysterols and control the expression of genes important for cholesterol uptake, efflux, transport, and excretion in multiple tissues. In addition to their role as cholesterol sensors, a number of studies have implicated LXRs in the modulation of innate and adaptive immune responses. Both through activation and repression mechanisms, LXRs regulate diverse aspects of inflammatory gene expression in macrophages. The ability of LXRs to coordinate metabolic and immune responses constitutes an attractive therapeutic target for the treatment of chronic inflammatory disorders. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.


Asunto(s)
Inflamación/fisiopatología , Macrófagos/fisiología , Receptores Nucleares Huérfanos/fisiología , Adaptación Fisiológica/inmunología , Animales , Apoptosis , Humanos , Inmunidad Innata/fisiología , Inflamación/genética , Mediadores de Inflamación/metabolismo , Receptores X del Hígado , Macrófagos/metabolismo , Factores de Transcripción/fisiología
15.
Immunity ; 31(2): 245-58, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19646905

RESUMEN

Effective clearance of apoptotic cells by macrophages is essential for immune homeostasis. The transcriptional pathways that allow macrophages to sense and respond to apoptotic cells are poorly defined. We found that liver X receptor (LXR) signaling was important for both apoptotic cell clearance and the maintenance of immune tolerance. Apoptotic cell engulfment activated LXR and thereby induced the expression of Mer, a receptor tyrosine kinase critical for phagocytosis. LXR-deficient macrophages exhibited a selective defect in phagocytosis of apoptotic cells and an aberrant proinflammatory response to them. As a consequence of these defects, mice lacking LXRs manifested a breakdown in self-tolerance and developed autoantibodies and autoimmune glomerulonephritis. Treatment with an LXR agonist ameliorated disease progression in a mouse model of lupus-like autoimmunity. Thus, activation of LXR by apoptotic cells engages a virtuous cycle that promotes their own clearance and couples engulfment to the suppression of inflammatory pathways.


Asunto(s)
Apoptosis/inmunología , Enfermedades Autoinmunes/inmunología , Proteínas de Unión al ADN/agonistas , Macrófagos/inmunología , Receptores Citoplasmáticos y Nucleares/agonistas , Bazo/inmunología , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Autoinmunidad/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/inmunología , Receptores X del Hígado , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nucleares Huérfanos , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/inmunología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/inmunología , Transducción de Señal/inmunología , Bazo/citología , Bazo/metabolismo , Tirosina Quinasa c-Mer
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA