Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters











Publication year range
1.
Trends Parasitol ; 40(6): 446-448, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38772757

ABSTRACT

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.


Subject(s)
Macrophages , Phagocytosis , Schistosoma mansoni , Animals , Macrophages/immunology , Macrophages/parasitology , Schistosoma mansoni/physiology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/immunology , Humans , Liver/parasitology , Liver/immunology , Axl Receptor Tyrosine Kinase , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/physiology , Efferocytosis
2.
Cell Death Dis ; 12(6): 611, 2021 06 12.
Article in English | MEDLINE | ID: mdl-34120143

ABSTRACT

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.


Subject(s)
Muscle, Skeletal/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Regeneration/genetics , c-Mer Tyrosine Kinase/physiology , Animals , Cell Survival/genetics , Cells, Cultured , Female , Male , Mice , Mice, Knockout , Muscle Development/genetics , Myoblasts/physiology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , c-Mer Tyrosine Kinase/genetics , Axl Receptor Tyrosine Kinase
4.
J Am Heart Assoc ; 9(18): e017071, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32865099

ABSTRACT

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.


Subject(s)
MAP Kinase Signaling System/physiology , Macrophages/pathology , Myocardial Infarction/pathology , Osteopontin/physiology , c-Mer Tyrosine Kinase/physiology , Animals , Disease Models, Animal , Flow Cytometry , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Osteopontin/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/physiology , c-Mer Tyrosine Kinase/metabolism
5.
J Nutr ; 150(5): 1167-1177, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32047914

ABSTRACT

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.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Hydroxybenzoates/administration & dosage , Animals , Anti-Inflammatory Agents , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Cells, Cultured , Dietary Supplements , Disease Models, Animal , Gene Expression/drug effects , Interleukin-1beta/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , NF-kappa B/metabolism , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/physiology
6.
J Neurosci ; 40(7): 1453-1482, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31896673

ABSTRACT

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.


Subject(s)
Hippocampus/cytology , Neurogenesis/physiology , Neurons/cytology , Phagocytosis/physiology , Animals , Apoptosis , Calcium Signaling , Cell Line, Tumor , Chromatin Assembly and Disassembly , Culture Media, Conditioned , Feedback, Physiological , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Hippocampus/growth & development , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia , Nerve Regeneration/physiology , Nerve Tissue Proteins/physiology , Receptors, Purinergic P2Y12/physiology , Transcriptome , c-Mer Tyrosine Kinase/physiology
7.
Cell Metab ; 31(2): 406-421.e7, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31839486

ABSTRACT

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.


Subject(s)
Liver Cirrhosis/metabolism , Liver/metabolism , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , c-Mer Tyrosine Kinase/physiology , ADAM17 Protein/metabolism , Animals , Cell Line , Chronic Disease , Humans , Liver/cytology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
8.
J Immunol Methods ; 473: 112636, 2019 10.
Article in English | MEDLINE | ID: mdl-31369739

ABSTRACT

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.


Subject(s)
Image Processing, Computer-Assisted/methods , Macrophages/immunology , Phagocytosis , Apoptosis , Humans , Jurkat Cells , c-Mer Tyrosine Kinase/antagonists & inhibitors , c-Mer Tyrosine Kinase/physiology
9.
Nat Rev Immunol ; 19(9): 539-549, 2019 09.
Article in English | MEDLINE | ID: mdl-31019284

ABSTRACT

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.


Subject(s)
Apoptosis , Macrophages/physiology , Animals , Antigens, Surface/physiology , Humans , Membrane Proteins/physiology , Milk Proteins , Phagocytosis , Phosphatidylserines/physiology , Receptor Protein-Tyrosine Kinases/physiology , c-Mer Tyrosine Kinase/physiology
10.
Inflammation ; 42(1): 113-123, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30143932

ABSTRACT

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.


Subject(s)
Inflammation/prevention & control , Macrophages/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , c-Mer Tyrosine Kinase/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cytokines/metabolism , Escherichia coli , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/microbiology , Mice , Phagocytosis/drug effects , RAW 264.7 Cells , RNA, Small Interfering/pharmacology , Suppressor of Cytokine Signaling 1 Protein/drug effects , Suppressor of Cytokine Signaling 1 Protein/metabolism , c-Mer Tyrosine Kinase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL