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












Base de datos
Intervalo de año de publicación
1.
Trends Parasitol ; 40(6): 446-448, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38772757

RESUMEN

Liebold et al. recently revealed how the identity of dying cells drives distinct changes to the macrophages which engulf and clear them, a process known as efferocytosis. During infection with the helminth Schistosoma mansoni, liver macrophages recapitulate these phenotypes, mediated by Axl/MerTK receptors and regulating egg burdens.


Asunto(s)
Macrófagos , Fagocitosis , Schistosoma mansoni , Animales , Macrófagos/inmunología , Macrófagos/parasitología , Schistosoma mansoni/fisiología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/inmunología , Humanos , Hígado/parasitología , Hígado/inmunología , Tirosina Quinasa del Receptor Axl , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Tirosina Quinasa c-Mer/metabolismo , Tirosina Quinasa c-Mer/fisiología , Eferocitosis
2.
Cell Death Dis ; 12(6): 611, 2021 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-34120143

RESUMEN

Skeletal muscle regeneration following injury results from the proliferation and differentiation of myogenic stem cells, called satellite cells, located beneath the basal lamina of the muscle fibers. Infiltrating macrophages play an essential role in the process partly by clearing the necrotic cell debris, partly by producing cytokines that guide myogenesis. Infiltrating macrophages are at the beginning pro-inflammatory, but phagocytosis of dead cells induces a phenotypic change to become healing macrophages that regulate inflammation, myoblast fusion and growth, fibrosis, vascularization and return to homeostasis. The TAM receptor kinases Mer and Axl are known efferocytosis receptors in macrophages functioning in tolerogenic or inflammatory conditions, respectively. Here we investigated their involvement in the muscle regeneration process by studying the muscle repair following cardiotoxin-induced injury in Mer-/- mice. We found that Axl was the only TAM kinase receptor expressed on the protein level by skeletal muscle and C2C12 myoblast cells, while Mer was the dominant TAM kinase receptor in the CD45+ cells, and its expression significantly increased during repair. Mer ablation did not affect the skeletal muscle weight or structure, but following injury it resulted in a delay in the clearance of necrotic muscle cell debris, in the healing phenotype conversion of macrophages and consequently in a significant delay in the full muscle regeneration. Administration of the TAM kinase inhibitor BMS-777607 to wild type mice mimicked the effect of Mer ablation on the muscle regeneration process, but in addition, it resulted in a long-persisting necrotic area. Finally, in vitro inhibition of TAM kinase signaling in C2C12 myoblasts resulted in decreased viability and in impaired myotube growth. Our work identifies Axl as a survival and growth receptor in the mouse myoblasts, and reveals the contribution of TAM kinase-mediated signaling to the skeletal muscle regeneration both in macrophages and in myoblasts.


Asunto(s)
Músculo Esquelético/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Regeneración/genética , Tirosina Quinasa c-Mer/fisiología , Animales , Supervivencia Celular/genética , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos/genética , Mioblastos/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa del Receptor Axl
4.
J Am Heart Assoc ; 9(18): e017071, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32865099

RESUMEN

Background We previously reported that osteopontin plays an essential role in accelerating both reparative fibrosis and clearance of dead cells (efferocytosis) during tissue repair after myocardial infarction (MI) and galectin-3hiCD206+ macrophages is the main source of osteopontin in post-MI heart. Interleukin-10- STAT3 (signal transducer and activator of transcription 3)-galectin-3 axis is essential for Spp1 (encoding osteopontin) transcriptional activation in cardiac macrophages after MI. Here, we investigated the more detailed mechanism responsible for functional maturation of osteopontin-producing macrophages. Methods and Results In post-MI hearts, Spp1 transcriptional activation occurred almost exclusively in MerTK (Mer tyrosine kinase)+ galectin-3hi macrophages. The induction of MerTK on galectin-3hi macrophages is essential for their functional maturation including efferocytosis and Spp1 transcriptional activity. MerTK+galectin-3hi macrophages showed a strong activation of both STAT3 and ERK (extracellular signal-regulated kinase). STAT3 inhibition suppressed the differentiation of osteopontin-producing MerTK+galectin-3hi macrophages, however, STAT3 activation was insufficient at inducing Spp1 transcriptional activity. ERK inhibition suppressed Spp1 transcriptional activation without affecting MerTK or galectin-3 expression. Concomitant activation of ERK is required for transcriptional activation of Spp1. In Il-10 knockout enhanced green fluorescent protein-Spp1 knock-in mice subjected to MI, osteopontin-producing macrophages decreased but did not disappear entirely. Interleukin-10 and macrophage colony-stimulating factor synergistically activated STAT3 and ERK and promoted the differentiation of osteopontin-producing MerTK+galectin-3hi macrophages in bone marrow-derived macrophages. Coadministration of anti-interleukin-10 plus anti-macrophage colony-stimulating factor antibodies substantially reduced the number of osteopontin-producing macrophages by more than anti-interleukin-10 antibody alone in post-MI hearts. Conclusions Interleukin-10 and macrophage colony-stimulating factor act synergistically to activate STAT3 and ERK in cardiac macrophages, which in turn upregulate the expression of galectin-3 and MerTK, leading to the functional maturation of osteopontin-producing macrophages.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos/patología , Infarto del Miocardio/patología , Osteopontina/fisiología , Tirosina Quinasa c-Mer/fisiología , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Macrófagos/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Osteopontina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/fisiología , Tirosina Quinasa c-Mer/metabolismo
5.
J Nutr ; 150(5): 1167-1177, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32047914

RESUMEN

BACKGROUND: Normalization of arterial inflammation inhibits atherosclerosis. The preventive role for protocatechuic acid (PCA) in early-stage atherosclerosis is well recognized; however, its therapeutic role in late-stage atherosclerosis remains unexplored. OBJECTIVE: We investigated whether PCA inhibits vulnerable atherosclerosis progression by normalizing arterial inflammation. METHODS: Thirty-wk-old male apolipoprotein E-deficient (Apoe-/-) mice with vulnerable atherosclerotic lesions in the brachiocephalic artery were fed the AIN-93G diet alone (control) or supplemented with 0.003% PCA (wt:wt) for 20 wk. Lesion size and composition, IL-1ß, and NF-κB in the brachiocephalic arteries, and serum lipid profiles, oxidative status, and proinflammatory cytokines (e.g., IL-1ß, monocyte chemoattractant protein-1, and serum amyloid A) were measured. Moreover, the effect of PCA on the inflammation response was evaluated in efferocytic macrophages from C57BL/6J mice. RESULTS: Compared with the control treatment, dietary PCA supplementation significantly reduced lesion size (27.5%; P < 0.05) and also improved lesion stability (P < 0.05) as evidenced by increased thin fibrous cap thickness (31.7%) and collagen accumulation (58.3%), reduced necrotic core size (37.6%) and cellular apoptosis (73.9%), reduced macrophage accumulation (45.1%), and increased vascular smooth muscle cell accumulation (51.5%). Moreover, PCA supplementation inhibited IL-1ß expression (53.7%) and NF-κB activation (64.4%) in lesions. However, PCA supplementation did not change serum lipid profiles, total antioxidant capacity, and inflammatory cytokines. In efferocytic macrophages, PCA at 0.5 and 1 µmol/L inhibited Il1b/IL-1ß mRNA (27.2-46.5%) and protein (29.2-49.6%) expression and NF-κB activation (67.0-80.3%) by upregulation of MER proto-oncogene tyrosine kinase (MERTK) and inhibition of mitogen-activated protein kinase 3/1 (MAPK3/1). Strikingly, the similar pattern of the MERTK and MAPK3/1 changes in lesional macrophages of mice after PCA intervention in vivo was recapitulated. CONCLUSION: PCA inhibits vulnerable lesion progression in mice, which might partially be caused by normalization of arterial inflammation by upregulation of MERTK and inhibition of MAPK3/1 in lesional macrophages.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Aterosclerosis/prevención & control , Hidroxibenzoatos/administración & dosificación , Animales , Antiinflamatorios , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Células Cultivadas , Suplementos Dietéticos , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , FN-kappa B/metabolismo , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/fisiología
6.
J Neurosci ; 40(7): 1453-1482, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31896673

RESUMEN

During adult hippocampal neurogenesis, most newborn cells undergo apoptosis and are rapidly phagocytosed by resident microglia to prevent the spillover of intracellular contents. Here, we propose that phagocytosis is not merely passive corpse removal but has an active role in maintaining neurogenesis. First, we found that neurogenesis was disrupted in male and female mice chronically deficient for two phagocytosis pathways: the purinergic receptor P2Y12, and the tyrosine kinases of the TAM family Mer tyrosine kinase (MerTK)/Axl. In contrast, neurogenesis was transiently increased in mice in which MerTK expression was conditionally downregulated. Next, we performed a transcriptomic analysis of the changes induced by phagocytosis in microglia in vitro and identified genes involved in metabolism, chromatin remodeling, and neurogenesis-related functions. Finally, we discovered that the secretome of phagocytic microglia limits the production of new neurons both in vivo and in vitro Our data suggest that microglia act as a sensor of local cell death, modulating the balance between proliferation and survival in the neurogenic niche through the phagocytosis secretome, thereby supporting the long-term maintenance of adult hippocampal neurogenesis.SIGNIFICANCE STATEMENT Microglia are the brain professional phagocytes and, in the adult hippocampal neurogenic niche, they remove newborn cells naturally undergoing apoptosis. Here we show that phagocytosis of apoptotic cells triggers a coordinated transcriptional program that alters their secretome, limiting neurogenesis both in vivo and in vitro In addition, chronic phagocytosis disruption in mice deficient for receptors P2Y12 and MerTK/Axl reduces adult hippocampal neurogenesis. In contrast, inducible MerTK downregulation transiently increases neurogenesis, suggesting that microglial phagocytosis provides a negative feedback loop that is necessary for the long-term maintenance of adult hippocampal neurogenesis. Therefore, we speculate that the effects of promoting engulfment/degradation of cell debris may go beyond merely removing corpses to actively promoting regeneration in development, aging, and neurodegenerative diseases.


Asunto(s)
Hipocampo/citología , Neurogénesis/fisiología , Neuronas/citología , Fagocitosis/fisiología , Animales , Apoptosis , Señalización del Calcio , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Medios de Cultivo Condicionados , Retroalimentación Fisiológica , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Hipocampo/crecimiento & desarrollo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores Purinérgicos P2Y12/fisiología , Transcriptoma , Tirosina Quinasa c-Mer/fisiología
7.
Cell Metab ; 31(2): 406-421.e7, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31839486

RESUMEN

Nonalcoholic steatohepatitis (NASH) is emerging as a leading cause of chronic liver disease. However, therapeutic options are limited by incomplete understanding of the mechanisms of NASH fibrosis, which is mediated by activation of hepatic stellate cells (HSCs). In humans, human genetic studies have shown that hypomorphic variations in MERTK, encoding the macrophage c-mer tyrosine kinase (MerTK) receptor, provide protection against liver fibrosis, but the mechanisms remain unknown. We now show that holo- or myeloid-specific Mertk targeting in NASH mice decreases liver fibrosis, congruent with the human genetic data. Furthermore, ADAM metallopeptidase domain 17 (ADAM17)-mediated MerTK cleavage in liver macrophages decreases during steatosis to NASH transition, and mice with a cleavage-resistant MerTK mutant have increased NASH fibrosis. Macrophage MerTK promotes an ERK-TGFß1 pathway that activates HSCs and induces liver fibrosis. These data provide insights into the role of liver macrophages in NASH fibrosis and provide a plausible mechanism underlying MERTK as a genetic risk factor for NASH fibrosis.


Asunto(s)
Cirrosis Hepática/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Tirosina Quinasa c-Mer/fisiología , Proteína ADAM17/metabolismo , Animales , Línea Celular , Enfermedad Crónica , Humanos , Hígado/citología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas
8.
J Immunol Methods ; 473: 112636, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31369739

RESUMEN

Macrophages are a diverse population of cells originating from the myeloid lineage, which form an important component of the innate immune system, helping to regulate immune response through secretion of pro/anti-inflammatory cytokines. However they also have an important homeostatic role - helping to remove cellular debris and apoptotic cells from the body (a phagocytic process known as efferocytosis). Here we describe a robust 384 well microplate based imaging assay, using apoptotic target cells for the specific quantification of efferocytosis in human primary monocyte derived macrophages. The methodology described allows for the assay to run in either fixed end-point or live-cell format (the former offering multiple morphological and intensity-based readouts, whilst the latter opens the possibility for future expansion of the methodology to encompass kinetic profiling). Within the methodology described we couple high content image acquisition (on the Cell Voyager 7000S) with multi-parametric image analysis - using Perkin Elmer Columbus combined with GeneData Screener.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Macrófagos/inmunología , Fagocitosis , Apoptosis , Humanos , Células Jurkat , Tirosina Quinasa c-Mer/antagonistas & inhibidores , Tirosina Quinasa c-Mer/fisiología
9.
Nat Rev Immunol ; 19(9): 539-549, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31019284

RESUMEN

Tissue macrophages rapidly recognize and engulf apoptotic cells. These events require the display of so-called eat-me signals on the apoptotic cell surface, the most fundamental of which is phosphatidylserine (PtdSer). Externalization of this phospholipid is catalysed by scramblase enzymes, several of which are activated by caspase cleavage. PtdSer is detected both by macrophage receptors that bind to this phospholipid directly and by receptors that bind to a soluble bridging protein that is independently bound to PtdSer. Prominent among the latter receptors are the MER and AXL receptor tyrosine kinases. Eat-me signals also trigger macrophages to engulf virus-infected or metabolically traumatized, but still living, cells, and this 'murder by phagocytosis' may be a common phenomenon. Finally, the localized presentation of PtdSer and other eat-me signals on delimited cell surface domains may enable the phagocytic pruning of these 'locally dead' domains by macrophages, most notably by microglia of the central nervous system.


Asunto(s)
Apoptosis , Macrófagos/fisiología , Animales , Antígenos de Superficie/fisiología , Humanos , Proteínas de la Membrana/fisiología , Proteínas de la Leche , Fagocitosis , Fosfatidilserinas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Tirosina Quinasa c-Mer/fisiología
10.
Am J Transplant ; 19(3): 674-685, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30133807

RESUMEN

Recipient infusion of donor apoptotic cells is an emerging strategy for inducing robust transplantation tolerance. Daily clearance of billions of self-apoptotic cells relies on homeostatic engagement of phagocytic receptors, in particular, receptors of the tyrosine kinase family TAM (Tyro3, Axl, and MerTK), to maintain self-tolerance. However, an outstanding question is if allogeneic apoptotic cells trigger the same receptor system for inducing allogeneic tolerance. Here, we employed allogeneic apoptotic splenocytes and discovered that the efferocytic receptor MerTK on recipient phagocytes is a critical mediator for transplantation tolerance induced by this strategy. Our findings indicate that the tolerogenic properties of allogeneic apoptotic splenocytes require MerTK transmission of intracellular signaling to suppress the production of inflammatory cytokine interferon α (IFN-α). We further demonstrate that MerTK is crucial for subsequent expansion of myeloid-derived suppressor cells and for promoting their immunomodulatory function, including maintaining graft-infiltrating CD4+ CD25+ Foxp3+ regulatory T cells. Consequently, recipient MerTK deficiency resulted in failure of tolerance by donor apoptotic cells, and this failure could be effectively rescued by IFN-α receptor blockade. These findings underscore the importance of the efferocytic receptor MerTK in mediating transplantation tolerance by donor apoptotic cells and implicate MerTK agonism as a promising target for promoting transplantation tolerance.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Rechazo de Injerto/etiología , Interferón Tipo I/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Tirosina Quinasa c-Mer/fisiología , Animales , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Experimental/terapia , Rechazo de Injerto/patología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Interferón Tipo I/metabolismo , Trasplante de Islotes Pancreáticos/efectos adversos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Donantes de Tejidos
11.
Inflammation ; 42(1): 113-123, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30143932

RESUMEN

Bacterial lipopolysaccharide (LPS) induces inflammatory response via toll-like receptor 4 (TLR4). However, this response must be strictly regulated because unbalanced overproduction of pro-inflammatory cytokines can lead to tissue damage and even be fatal. Herein, we explore whether Mer receptor tyrosine kinase (MerTK) regulates Escherichia coli (E. coli) LPS-induced inflammation and mediates phagocytosis of E. coli by macrophages. The results showed that LPS activated TLR4 signaling pathway and induced MerTK pathway in RAW264.7 macrophages, including suppressor of cytokine signaling1 (SOCS1). Preincubation with MerTK-specific blocking antibody (MerTK-Ab) markedly suppressed LPS-induced expression of phosphorylated MerTK, while further promoted LPS-induced production of TNF-α, IL-6, and IL-1ß as well as phosphorylation of IκB-α and p65. Likewise, MerTK-Ab prevented LPS-induced SOCS1 expression. Furthermore, LPS-induced production of pro-inflammatory cytokines and activation of NF-κB were increased by transfection with SOCS1 siRNA. Additionally, we demonstrated that MerTK was dispensable in phagocytosis of E. coli by RAW264.7 or peritoneal macrophages. Collectively, these results indicate that MerTK downregulates LPS-induced inflammation through SOCS1 protein without affecting phagocytosis of E. coli in macrophages.


Asunto(s)
Inflamación/prevención & control , Macrófagos/inmunología , Proteína 1 Supresora de la Señalización de Citocinas/genética , Tirosina Quinasa c-Mer/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Citocinas/metabolismo , Escherichia coli , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/microbiología , Ratones , Fagocitosis/efectos de los fármacos , Células RAW 264.7 , ARN Interferente Pequeño/farmacología , Proteína 1 Supresora de la Señalización de Citocinas/efectos de los fármacos , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Tirosina Quinasa c-Mer/inmunología
12.
Adv Exp Med Biol ; 1074: 577-583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721990

RESUMEN

MerTK is required for photoreceptor outer segment (POS) phagocytosis by retinal pigment epithelial (RPE) cells, a diurnal function essential for vision maintenance. In vivo, MerTK is stimulated at the time of the phagocytic peak through an intracellular signaling pathway. However, MerTK ligands Gas6 and Protein S are expressed in both RPE cells and photoreceptors, and at least one of them required for phagocytosis to occur. Still, their exact role in the retina was not clear until recently. This review combines results from different studies to shed the light on a tissue-specific regulation of MerTK function by its ligands. Indeed, with opposite effects on RPE phagocytosis and changes in their expression levels around the time of POS uptake, Gas6 and Protein S may contribute to the tight control of the acute phagocytic peak in the retina.


Asunto(s)
Apoptosis/fisiología , Proteínas del Ojo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Fagocitosis/fisiología , Proteína S/fisiología , Retina/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Animales , Células Cultivadas , Ritmo Circadiano , Activación Enzimática , Humanos , Ligandos , Macrófagos/metabolismo , Ratones , Ratas , Retina/citología , Segmento Externo de la Célula en Bastón/metabolismo , Transducción de Señal/fisiología , Tirosina Quinasa c-Mer/deficiencia , Tirosina Quinasa c-Mer/fisiología
13.
Front Immunol ; 9: 742, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29706963

RESUMEN

Objective: Rheumatoid arthritis (RA) is a chronic and progressive joint disease. It appears that anti-inflammatory feedback mechanisms that could restrain joint inflammation and restore homeostasis are insufficient to perform this control. In this study, we investigated the contribution of the MER tyrosine kinase-mediated anti-inflammatory response on arthritis and whether targeting MER could be a valid approach to treat RA. Methods: KRN serum transfer arthritis (KRN STA) was induced in either Mertk-deficient mice or in mice that adenovirally overexpressed Pros1. Human synovial micromasses were treated with MER-specific antibodies or PROS1. Collagen-induced arthritis (CIA) mice were treated with MER-specific agonistic antibodies or by viral overexpression of Pros1. Results: Mertk-/- mice showed exacerbated arthritis pathology, whereas Pros1 overexpression diminished joint pathology in KRN STA. Human synovial micromasses challenged with MER-specific antibodies enhanced the secretion of inflammatory cytokines, whereas stimulating MER with PROS1 reduced the secretion of these cytokines, confirming the protective role of MER. Next, we treated CIA mice with MER-specific agonistic antibodies, and this unexpectedly resulted in exacerbated arthritis pathology. This was associated with increased numbers of apoptotic cells in their knee joints and higher serum levels of interleukin (IL)-16C, a cytokine released by secondary necrotic neutrophils. Apoptotic cell numbers and IL-16C levels were enhanced during arthritis in Mertk-/- mice and reduced in Pros1-overexpressing mice. Conclusion: MER plays a protective role during joint inflammation and activating MER by its ligand PROS1 ameliorates disease. Treatment of mice with MER receptor agonistic antibodies is deleterious due to its counterproductive effect of blocking efferocytosis in the arthritic joint.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Proteínas Portadoras/fisiología , Tirosina Quinasa c-Mer/fisiología , Animales , Artritis Experimental/patología , Artritis Reumatoide/patología , Proteínas de Unión al Calcio , Línea Celular , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Membrana Sinovial/inmunología
14.
Pharmacol Ther ; 188: 97-117, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29514053

RESUMEN

Tyro3, Axl, and Mertk, referred to as the TAM family of receptor tyrosine kinases, are instrumental in maintaining cell survival and homeostasis in mammals. TAM receptors interact with multiple signaling molecules to regulate cell migration, survival, phagocytosis and clearance of metabolic products and cell debris called efferocytosis. The TAMs also function as rheostats to reduce the expression of proinflammatory molecules and prevent autoimmunity. All three TAM receptors are activated in a concentration-dependent manner by the vitamin K-dependent growth arrest-specific protein 6 (Gas6). Gas6 and the TAMs are abundantly expressed in the nervous system. Gas6, secreted by neurons and endothelial cells, is the sole ligand for Axl. ProteinS1 (ProS1), another vitamin K-dependent protein functions mainly as an anti-coagulant, and independent of this function can activate Tyro3 and Mertk, but not Axl. This review will focus on the role of the TAM receptors and their ligands in the nervous system. We highlight studies that explore the function of TAM signaling in myelination, the visual cortex, neural cancers, and multiple sclerosis (MS) using Gas6-/- and TAM mutant mice models.


Asunto(s)
Sistema Nervioso/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Tirosina Quinasa c-Mer/fisiología , Animales , Proteínas Sanguíneas/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ligandos , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Proteína S , Transducción de Señal , Vitamina K/fisiología , Tirosina Quinasa del Receptor Axl
15.
Pharmacol Ther ; 188: 12-25, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29444453

RESUMEN

Atherosclerosis is a dynamic and progressive inflammatory process in the intimal layer of large and medium-sized arteries, and it is the major contributor to the atherosclerotic cardiovascular disease (ACVD), the leading cause of death worldwide. In an atherosclerotic plaque, phagocytosis of apoptotic cells occurs through an intricate process designated efferocytosis. Defective efferocytosis has emerged as a causal factor in the etiopathogenesis of atherosclerosis and its progression into overt ACVD. Both specialized phagocytes (macrophages and dendritic cells) and non-specialized cells with phagocytic capabilities (smooth muscle and endothelial cells) are involved in the efferocytotic process. Moreover, several signaling and regulatory molecules are involved in the different steps of efferocytosis, and they include "Find-Me" signals (lysophosphatidylcholine), "Eat-Me" signals [phosphatidylserine, Mer tyrosine kinase (MerTK), and milk fat globule-EGF factor 8], and "Don't Eat-Me" signals [cluster of differentiation 47 (CD47)]. Regulation of efferocytosis is in a close nexus with inflammation, the key component in atherosclerosis. The predominance of pro-inflammatory and anti-inflammatory molecules plays a crucial role in lesion progression and regression, respectively. Polarization of macrophages towards the M1 phenotype causes them to secrete proinflammatory cytokines, while polarization towards the M2 phenotype causes them to secrete of anti-inflammatory cytokines, including interleukin-10 and transforming growth factor ß, so tending to shift the balance towards resolution of the inflammation. Dysfunction of any regulatory signal may cause expansion of the necrotic core of an atherosclerotic plaque with ensuing conversion of the plaque into an unstable plaque with an increased susceptibility to rupture and to atherothrombotic complication. In this review we aim at elucidating the determinant factors and pathways of efferocytosis which can be considered as potential novel targets when striving to develop more personalized and efficient treatment regimens for patients with ACVD.


Asunto(s)
Aterosclerosis/etiología , Fagocitosis/fisiología , Antígenos de Superficie/fisiología , Apoptosis , Antígeno CD47/fisiología , Calreticulina/fisiología , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Macrófagos/fisiología , Proteínas de la Leche , Tirosina Quinasa c-Mer/fisiología
17.
J Leukoc Biol ; 103(4): 709-717, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29350876

RESUMEN

Endotoxin induces a variety of proinflammatory mediators and plays a crucial role in kidney inflammation. The receptor tyrosine kinase, Mer, diminishes renal inflammation by attenuating inflammatory responses. We previously reported that Mer is predominantly expressed on glomerular endothelial cells (GECs) and that Mer deficiency is associated with increased renal inflammation when mice are challenged with nephrotoxic serum. We consequently hypothesized that Mer signaling down-regulates LPS-driven inflammatory responses in GECs. To test this hypothesis, primary GECs were isolated from the kidneys of Mer-KO and wild-type (WT) control mice. LPS treatment induced Akt and STAT3 activation along with Bcl-xl up-regulation in WT GECs; these responses were all increased in Mer-deficient GECs. In addition, STAT1 and ERK1/2 up-regulation and activation were observed in Mer-KO GECs exposed to LPS. In contrast, expression of the inhibitory signaling molecule, suppressor of cytokine signaling-3 (SOCS-3), was much higher in LPS-stimulated WT than Mer-deficient GECs. Deficiency of Mer was also associated with significantly increased NF-κB expression and activation. These observations indicate that Mer functions as an intrinsic feedback inhibitor of inflammatory mediator-driven immune responses in GECs during kidney injury and suggest a new therapeutic strategy for glomerular diseases.


Asunto(s)
Células Endoteliales/inmunología , Regulación de la Expresión Génica , Inflamación/prevención & control , Glomérulos Renales/inmunología , Tirosina Quinasa c-Mer/fisiología , Animales , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Inflamación/inducido químicamente , Inflamación/metabolismo , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal
18.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L69-L82, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935638

RESUMEN

Microparticles are a newly recognized class of mediators in the pathophysiology of lung inflammation and injury, but little is known about the factors that regulate their accumulation and clearance. The primary objective of our study was to determine whether alveolar macrophages engulf microparticles and to elucidate the mechanisms by which this occurs. Alveolar microparticles were quantified in bronchoalveolar fluid of mice with lung injury induced by LPS and hydrochloric acid. Microparticle numbers were greatest at the peak of inflammation and declined as inflammation resolved. Isolated, fluorescently labeled particles were placed in culture with macrophages to evaluate ingestion in the presence of endocytosis inhibitors. Ingestion was blocked with cytochalasin D and wortmannin, consistent with a phagocytic process. In separate experiments, mice were treated intratracheally with labeled microparticles, and their uptake was assessed though microscopy and flow cytometry. Resident alveolar macrophages, not recruited macrophages, were the primary cell-ingesting microparticles in the alveolus during lung injury. In vitro, microparticles promoted inflammatory signaling in LPS primed epithelial cells, signifying the importance of microparticle clearance in resolving lung injury. Microparticles were found to have phosphatidylserine exposed on their surfaces. Accordingly, we measured expression of phosphatidylserine receptors on macrophages and found high expression of MerTK and Axl in the resident macrophage population. Endocytosis of microparticles was markedly reduced in MerTK-deficient macrophages in vitro and in vivo. In conclusion, microparticles are released during acute lung injury and peak in number at the height of inflammation. Resident alveolar macrophages efficiently clear these microparticles through MerTK-mediated phagocytosis.


Asunto(s)
Lesión Pulmonar Aguda/fisiopatología , Micropartículas Derivadas de Células/fisiología , Inflamación/patología , Macrófagos Alveolares/fisiología , Fagocitosis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa c-Mer/fisiología , Lesión Pulmonar Aguda/metabolismo , Animales , Apoptosis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Tirosina Quinasa del Receptor Axl
19.
J Immunol ; 199(12): 4001-4015, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29118245

RESUMEN

Mer tyrosine kinase (Mer) signaling maintains immune tolerance by clearing apoptotic cells (ACs) and inducing immunoregulatory signals. We previously showed that Mer-deficient mice (Mer-/-) have increased germinal center (GC) responses, T cell activation, and AC accumulation within GCs. Accumulated ACs in GCs can undergo necrosis and release self-ligands, which may influence the outcome of a GC response and selection. In this study, we generated Mer-/- mice with a global MyD88, TLR7, or TLR9 deficiency and cell type-specific MyD88 deficiency to study the functional correlation between Mer and TLRs in the development of GC responses and autoimmunity. We found that GC B cell-intrinsic sensing of self-RNA, but not self-DNA, released from dead cells accumulated in GCs drives enhanced GC responses in Mer-/- mice. Although self-ligands directly affect GC B cell responses, the loss of Mer in dendritic cells promotes enhanced T cell activation and proinflammatory cytokine production. To study the impact of Mer deficiency on the development of autoimmunity, we generated autoimmune-prone B6.Sle1b mice deficient in Mer (Sle1bMer-/-). We observed accelerated autoimmunity development even under conditions where Sle1bMer-/- mice did not exhibit increased AC accumulation in GCs compared with B6.Sle1b mice, indicating that Mer immunoregulatory signaling in APCs regulates B cell selection and autoimmunity. We further found significant expansion, retention, and class-switching of autoreactive B cells in GCs under conditions where ACs accumulated in GCs of Sle1bMer-/- mice. Altogether, both the phagocytic and immunomodulatory functions of Mer regulate GC responses to prevent the development of autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Centro Germinal/inmunología , Autotolerancia/fisiología , Tirosina Quinasa c-Mer/fisiología , Animales , Presentación de Antígeno , Apoptosis , Subgrupos de Linfocitos B/inmunología , Femenino , Inmunización , Cambio de Clase de Inmunoglobulina , Riñón/patología , Masculino , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , ARN/inmunología , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 9/deficiencia , Tirosina Quinasa c-Mer/deficiencia , Tirosina Quinasa c-Mer/genética
20.
Inflammation ; 40(5): 1543-1552, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28528507

RESUMEN

Mer receptor tyrosine kinase (MerTK) expressed in macrophages is essential for phagocytosis of apoptotic cells. Here, we investigate whether MerTK is involved in the phagocytosis of Staphylococcus aureus (S. aureus) and regulation of staphylococcal lipoteichoic acid (LTA)-induced inflammatory response in macrophages. We found that stimulating RAW264.7 macrophages with S. aureus activated multiple signaling pathways including toll-like receptor 2 (TLR2), scavenger receptor A (SR-A), and MerTK. Meanwhile, S. aureus stimulation also induced activation of proteins focal adhesion kinase (FAK) and Rac1, which are related to phagocytosis. Pretreatment with a specific Mer-blocking antibody significantly inhibited S. aureus-induced phosphorylation of MerTK, while it had no effect on S. aureus-induced activation of FAK and Rac1. Moreover, by confocal laser microscope, we observed that the antibody blockade of MerTK had little impact on the phagocytosis of S. aureus by RAW264.7 macrophages. Additionally, pretreatment with this antibody further promoted LTA-induced phosphorylation of nuclear factor κB (NF-κB) p65 subunit and production of pro-inflammatory cytokines, such as TNF-α, IL-6, IL-1ß, and macrophage inflammatory protein-2 (MIP-2). Collectively, these results suggest that MerTK does not play an essential role in the phagocytosis of S. aureus but attenuates inflammation induced by staphylococcal LTA through blocking NF-κB activation.


Asunto(s)
Inflamación/prevención & control , Staphylococcus aureus/inmunología , Tirosina Quinasa c-Mer/fisiología , Animales , Inflamación/inducido químicamente , Lipopolisacáridos/efectos adversos , Ratones , FN-kappa B/antagonistas & inhibidores , Fagocitosis , Células RAW 264.7 , Staphylococcus aureus/patogenicidad , Ácidos Teicoicos/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...