scholarly journals IRF5 Is a Specific Marker of Inflammatory MacrophagesIn Vivo

2013 ◽  
Vol 2013 ◽  
pp. 1-9 ◽  
Author(s):  
Miriam Weiss ◽  
Katrina Blazek ◽  
Adam J. Byrne ◽  
Dany P. Perocheau ◽  
Irina A. Udalova

Macrophages are an integral part of the innate immune system and key players in pathogen clearance and tissue remodelling. Both functions are accomplished by a pivotal network of different macrophage subtypes, including proinflammatory M1 and anti-inflammatory M2 macrophages. Previously, our laboratory identified the transcription factor interferon regulatory factor 5 (IRF5) as the master regulator of the M1 macrophage polarisation. IRF5 was found to be highly expressed in human M1 compared to M2 macrophages. Furthermore, IRF5 dictates the expression of proinflammatory genes such asIL12bandIL23awhilst repressing anti-inflammatory genes likeIL10. Here we show that murine bone marrow derived macrophages differentiatedin vitrowith GM-CSF are also characterised by high levels of IRF5 mRNA and protein and express proinflammatory cytokines upon LPS stimulation. These macrophages display characteristic expression of M1-marker MHC II but lack the M2-marker CD206. Significantly, we develop intracellular staining of IRF5- expressing macrophages and utilise it to recapitulate thein vitroresults in anin vivomodel of antigen-induced arthritis, emphasising their physiological relevance. Thus, we establish the species-invariant role of IRF5 in controlling the inflammatory macrophage phenotype bothin vitroand inin vivo.

Author(s):  
С.В. Калиш ◽  
С.В. Лямина ◽  
А.А. Раецкая ◽  
И.Ю. Малышев

Цель исследования. Репрограммирование М1 фенотипа макрофагов с ингибированными факторами транскрипции М2 фенотипа STAT3, STAТ6 и SMAD и оценка их влияния на развитие карциномы Эрлиха (КЭ) in vitro и in vivo. Методика. Рост опухоли иницировали in vitro путем добавления клеток КЭ в среду культивирования RPMI-1640 и in vivo путем внутрибрюшинной инъекции клеток КЭ мышам. Результаты. Установлено, что M1макрофаги и in vitro, и in vivo оказывают выраженный противоопухолевый эффект, который превосходит антиопухолевые эффекты М1, M1, M1 макрофагов и цисплатина. Заключение. М1 макрофаги с ингибированными STAT3, STAT6 и/или SMAD3 эффективно ограничивают рост опухоли. Полученные данные обосновывают разработку новой технологии противоопухолевой клеточной терапии. Objective. Reprogramming of M1 macrophage phenotype with inhibited M2 phenotype transcription factors, such as STAT3, STAT6 and SMAD and assess their impact on the development of Ehrlich carcinoma (EC) in vitro and in vivo . Methods. Tumor growth in vitro was initiated by addition of EC cells in RPMI-1640 culture medium and in vivo by intraperitoneal of EC cell injection into mice. Results. It was found that M1 macrophages have a pronounced anti-tumor effect in vitro , and in vivo , which was greater than anti-tumor effects of M1, M1, M1 macrophages and cisplatin. Conclusion. M1 macrophages with inhibited STAT3, STAT6 and/or SMAD3 effectively restrict tumor growth. The findings justify the development of new anti-tumor cell therapy technology.


2011 ◽  
Vol 140 (5) ◽  
pp. S-19
Author(s):  
Michelle Taylor ◽  
Vandana Gambhir ◽  
Curtis Noordhof ◽  
Oliver Jones ◽  
Shu-Mei He ◽  
...  

2019 ◽  
Vol 133 (7) ◽  
pp. 869-884 ◽  
Author(s):  
Xianglan Liu ◽  
Ruoxi Zhang ◽  
Jingbo Hou ◽  
Jian Wu ◽  
Maomao Zhang ◽  
...  

Abstract Background: Early strut coverage after sirolimus-eluting stent (SES) implantation is associated with the activation of inflammation, but the underlying mechanisms are not completely understood. The present study aimed to identify the relationship between the anti-inflammatory cytokine interleukin (IL) 35 (IL-35) and early strut coverage in vivo and in vitro. Methods: We utilized a retrospective study design to measure IL-35 levels in 68 stents from 68 patients with coronary artery disease and recorded serial optical coherence tomography (OCT) images (0 and 3 months) to assess stent endothelialization. The mechanism underlying the regulatory effects of IL-35 on macrophages and human umbilical vein endothelial cells (HUVECs) was also investigated. SESs were surgically implanted into the right common carotid arteries of 200 male New Zealand White rabbits receiving intravenous injections of IL-35 or a placebo. Results: At the 3-month OCT evaluation, complete endothelium coverage was correlated with IL-35 levels. IL-35 induced the activation of an anti-inflammatory M2-like macrophage phenotype by targeting the signal transducer and activators of transcription (STAT)1/4 signalling pathway, and IL-35-treated macrophages induced endothelial proliferation and alleviated endothelial dysfunction. IL-35-treated New Zealand White rabbits with implanted SESs showed lower percentages of cross-sections with an uncovered strut, elevated mean neointimal hyperplasia (NIH) thickness, and inhibited inflammatory responses. Conclusions: We investigated the effect of IL-35 expression on early stent endothelialization in vivo and in vitro and identified a crucial role for IL-35 in inducing the activation of an anti-inflammatory M2-like macrophage phenotype. The present study highlights a new therapeutic strategy for early stent endothelialization.


2017 ◽  
Vol 37 (suppl_1) ◽  
Author(s):  
Paul Mueller ◽  
Lin Zhu ◽  
Illaria Giunzioni ◽  
Hagai Tavori ◽  
John M Stafford ◽  
...  

We previously showed that mice lacking macrophage LDL receptor-related protein 1 (LRP1) undergo accelerated lesion formation due to increased apoptosis, decreased efferocytosis, and enhanced macrophage transformation into the inflammatory M1 phenotype. In vitro, LRP1-deficient macrophages (MΦLRP1 -/- ) show enhanced plasticity with exaggerated polarization towards either the inflammatory M1- or the anti-inflammatory M2-phenotype depending on the stimulant (LPS or IL-4, respectively). During atherosclerosis regression, the M2:M1 macrophage ratio increases as lesion M1 macrophages egress and inflammation resolves. Thus, we hypothesize that atherosclerosis regression is accelerated in MΦLRP1 -/- mice via enhanced macrophage M2 polarization and CCR7-dependent M1 macrophage egress. ApoE-/- mice on high fat diet for 12 weeks were reconstituted with bone marrow from wildtype (WT) or MΦLRP1 -/- mice and then placed on chow diet for 8 weeks. In this model, apoE is reintroduced into circulation to correct the hyperlipidemia and induce regression of atherosclerotic lesions. A cohort of apoE -/- mice reconstituted with apoE -/- bone marrow served as baseline controls. Lesions in both WT and MΦLRP1 -/- mice regressed relative to controls (11% and 22%, respectively; p<0.05), but MΦLRP1 -/- lesions were 13% smaller than those of WT mice (p<0.05). LRP1 deletion increased M2 transformation of macrophages and a higher M2:M1 macrophage ratio (p<0.01) in the plaque. MΦLRP1 -/- lesions contained 36% fewer M1 macrophages compared to WT (p<0.01). In vivo studies of reverse cholesterol transport (RCT) revealed that MΦLRP1 -/- have a 1.4-fold higher RCT compared to WT mice (p<0.01). MΦLRP1 -/- lesions contained 2.5-fold more CCR7 + macrophages relative to WT lesions (p<0.01), and in our in vivo egress assay 4.6-fold more CCR7 + macrophages were found in mediastinal lymph nodes. In vitro , M1-differentiated MΦLRP1 -/- macrophages expressed 1.6-fold higher Ccr7 mRNA compared to WT controls (p<0.01). Thus, the absence of macrophage LRP1 accelerates atherosclerosis regression due to enhanced transformation of macrophages into an anti-inflammatory M2 phenotype, increased cholesterol efflux, and increased CCR7-driven egress of M1 macrophages from lesions.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Yu Zhao ◽  
Xiao liang Zhang ◽  
Bicheng Liu ◽  
Lilach Lerman

Abstract Background and Aims Tumor necrosis factor-α-induced gene/protein (TSG)-6 is a key factor influencing mesenchymal stem cells (MSCs) immunomodulatory properties, but its renoprotective efficacy is unknown. Using a novel swine model of renal artery stenosis (RAS) complicated by metabolic syndrome (Mets), we assessed the therapeutic effects of adipose tissue-derived MSCs-produced TSG-6 and mechanisms underlying the immunomodulatory properties of MSCs. Method Five groups of pigs (n=6 each) were studied after 16 weeks of diet-induced Mets and unilateral RAS (Mets+RAS), either untreated or treated 4 weeks earlier with a single intra-renal delivery of autologous posrcine adipose tissue-derived MSCs (pMSC). Lean, Mets, and RAS shams served as controls. We studied renal function in vivo (using CT imaging) and kidney histopathology and macrophage phenotype ex vivo. In vitro, TSG-6 levels were measured in conditioned media of human MSCs (hMSCs) incubated with or without TNF-α. Additionally, levels of the tubular injury marker LDH were measured in conditioned media after co-culturing macrophages with injured HK-2 cells (achieved by TNF-α and antimycin-A, AMA) with or without addition of TSG-6. The effects of TSG-6 on macrophage phenotype (M1/M2), adhesion, and migration capability were determined. Results Mets+RAS pigs showed increased renal M1 macrophages and renal vein TNF-α levels. After p-MSCs delivery, renal vein TSG-6 increased and TNF-α decreased, M1 macrophage switched to M2 (Fig. A),, renal function improved, and fibrosis alleviated. In vitro, TNF-α increased TSG-6 secretion by h-MSCs. TSG-6 decreased LDH release from injured HK-2, induced a macrophage phenotypic switch from M1 to M2 (Fig. B), and reduced M1 macrophage adhesion and migration (Fig. C). Conclusion TNF-α-induced TSG-6 release from MSCs in vivo and in vitro may decrease renal tubular cells injury, which is associated with and may be at least in part mediated by regulating macrophage function and phenotype.


Nutrients ◽  
2018 ◽  
Vol 10 (12) ◽  
pp. 1991 ◽  
Author(s):  
Yuji Morita ◽  
Yukihiro Miwa ◽  
Kenta Jounai ◽  
Daisuke Fujiwara ◽  
Toshihide Kurihara ◽  
...  

Age-related macular degeneration and retinitis pigmentosa are leading causes of blindness and share a pathological feature, which is photoreceptor degeneration. To date, the lack of a potential treatment to prevent such diseases has raised great concern. Photoreceptor degeneration can be accelerated by excessive light exposure via an inflammatory response; therefore, anti-inflammatory agents would be candidates to prevent the progress of photoreceptor degeneration. We previously reported that a lactic acid bacterium, Lactobacillus paracasei KW3110 (L. paracasei KW3110), activated macrophages suppressing inflammation in mice and humans. Recently, we also showed that intake of L. paracasei KW3110 could mitigate visual display terminal (VDT) load-induced ocular disorders in humans. However, the biological mechanism of L. paracasei KW3110 to retain visual function remains unclear. In this study, we found that L. paracasei KW3110 activated M2 macrophages inducing anti-inflammatory cytokine interleukin-10 (IL-10) production in vitro using bone marrow-derived M2 macrophages. We also show that IL-10 gene expression was significantly increased in the intestinal immune tissues 6 h after oral administration of L. paracasei KW3110 in vivo. Furthermore, we demonstrated that intake of L. paracasei KW3110 suppressed inflammation and photoreceptor degeneration in a murine model of light-induced retinopathy. These results suggest that L. paracasei KW3110 may have a preventive effect against degrative retinal diseases.


2021 ◽  
Vol 2021 ◽  
pp. 1-19
Author(s):  
Shaoxi Yan ◽  
Mo Zhou ◽  
Xiaoyun Zheng ◽  
Yuanyuan Xing ◽  
Juan Dong ◽  
...  

Inflammation causes tissue damage and promotes ventricular remodeling after myocardial infarction (MI), and the infiltration and polarization of macrophages play an important role in regulating inflammation post-MI. Here, we investigated the anti-inflammatory function of curcumin after MI and studied its relationship with macrophage polarization. In vivo, curcumin not only attenuated ventricular remodeling 3 months after MI but also suppressed inflammation during the first 7 days post-MI. Importantly, the results of qPCR and immunochemistry showed that curcumin decreased M1 (iNOS, CCL2, and CD86) but increased M2 macrophage (Arg1, CD163, and CD206) marker expression in the myocardium of MI mice during the first 7 days post-MI. And flow cytometry analysis indicated that curcumin suppressed M1 (CD45+Gr-1-CD11b+iNOS+ cells) but enhanced M2 macrophage (CD45+Gr-1-CD11b+Arg+ cells) expansion in the myocardium of MI mice during the first 7 days post-MI. In vitro, curcumin decreased LPS/IFNγ-elevated M1 macrophage marker (iNOS and CD86) expression and the proportion of M1 macrophages (iNOS+F4/80+ cells) but increased LPS/IFNγ-suppressed M2 macrophage marker (Arg1 and CD206) expression and the proportion of M2 macrophages (Arg1+F4/80+ cells). In addition, curcumin modulates M1/M2 macrophage polarization partly via AMPK. In conclusion, curcumin suppressed the MI-induced inflammation by modulating macrophage polarization partly via the AMPK pathway.


2020 ◽  
Vol 8 ◽  
pp. 100027 ◽  
Author(s):  
Aysegul Dede Eren ◽  
Ravi Sinha ◽  
Egemen Deniz Eren ◽  
Yuan Huipin ◽  
Sultan Gulce-Iz ◽  
...  

2019 ◽  
Author(s):  
Yong Zhu ◽  
Qiong Li ◽  
Weiping Kuang ◽  
Jun Lu ◽  
Qin Wang ◽  
...  

Abstract Background : Increasing evidence has demonstrated that circular RNAs (circRNAs) participate in epileptogenesis, but the expression profile and role of circRNAs in epilepsy remain unknown. A circRNA microarray was performed to examine epilepsy-related circRNAs. Bioinformatics analyses, luciferin reporter experiments and real-time quantitative PCR (Rt-qPCR) in vitro experiments were performed to demonstrate the mechanism of circRNA-mediated gene regulation of the microglial phenotype under epileptic conditions. Then, to further confirm the effect of circRNAs on nerve damage in the hippocampus, a mouse model of epilepsy was established by intraperitoneal injection of lithium chloride and pilocarpine. Results: The data indicated that 364 circRNAs were differentially expressed comparing epilepsy and control tissues. In particular, mmu_circ_0000335 expression was significantly downregulated in epileptic mice which was confirmed by Rt-qPCR. Overexpression of mmu_circ_0000335 promoted BV2 cell transformation into the M2 macrophage phenotype by increasing expression of CD206, Arg1, Ym1 and IL-10 while decreasing M1 macrophage markers IL-1β, IL-6, TNF-α and IFN-γ expressions under epileptic conditions. mmu_circ_0000335 expression triggered upregulation of Suppressor of Cytokine Signaling 1 (SOCS1) by decreasing miR-19b-3p levels, as determined by luciferase reporter assay. In vivo studies found that mmu_circ_0000335 overexpression decreased epilepsy-induced neural cell apoptosis in the hippocampus by reducing inflammatory cytokine expression. Immunofluorescence detection showed that mmu_circ_0000335 overexpression promoted microglial transformation into the M2 phenotype which had an anti-inflammatory effect. Conclusions: These results collectively indicated that mmu_circ_0000335 was involved in epilepsy progression by functioning as a miR-19b-3p sponge to enhance SOCS1 expression. Thus, mmu_circ_0000335 may be a candidate therapeutic target for epilepsy patients.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Ying Zhang ◽  
Conghui Zhang ◽  
Lixi Li ◽  
Xinjun Liang ◽  
Peng Cheng ◽  
...  

AbstractInflammation plays a crucial role in the occurrence and development of renal fibrosis, which ultimately results in end-stage renal disease (ESRD). There is new focus on lymphangiogenesis in the field of inflammation. Recent studies have revealed the association between lymphangiogenesis and renal fibrosis, but the source of lymphatic endothelial cells (LECs) is not clear. It has also been reported that macrophages are involved in lymphangiogenesis through direct and indirect mechanisms in other tissues. We hypothesized that there was a close relationship between macrophages and lymphatic endothelial progenitor cells in renal fibrosis. In this study, we demonstrated that lymphangiogenesis occurred in a renal fibrosis model and was positively correlated with the degree of fibrosis and macrophage infiltration. Compared to resting (M0) macrophages and alternatively activated (M2) macrophages, classically activated (M1) macrophages predominantly transdifferentiated into LECs in vivo and in vitro. VEGF-C further increased M1 macrophage polarization and transdifferentiation into LECs by activating VEGFR3. It was suggested that VEGF-C/VEGFR3 pathway activation downregulated macrophage autophagy and subsequently regulated macrophage phenotype. The induction of autophagy in macrophages by rapamycin decreased M1 macrophage polarization and differentiation into LECs. These results suggested that M1 macrophages promoted lymphangiogenesis and contributed to newly formed lymphatic vessels in the renal fibrosis microenvironment, and VEGF-C/VEGFR3 signaling promoted macrophage M1 polarization by suppressing macrophage autophagy and then increased the transdifferentiation of M1 macrophages into LECs.


Sign in / Sign up

Export Citation Format

Share Document