Macrophage reprogramming

巨噬细胞重编程
  • 文章类型: Journal Article
    普氏粪杆菌(F.prausnitzii)因其对人类的各种肠道和肠外益处而得到认可。并且普氏F.prausnitzii的减少与克罗恩病(CD)患者肠纤维化的风险增加有关。在这项研究中,口服活普氏弧菌或其细胞外囊泡(FEV)可以显着减轻重复给药DSS诱导的小鼠纤维化的严重程度。体外实验表明,FEV能够引导外周血单核细胞(PBMC)向M2b巨噬细胞表型的极化,与抗纤维化活性有关。发现FEV的这种作用在促进促纤维化M1/M2a/M2c巨噬细胞发育的各种条件下是稳定的。进行蛋白质组学和RNA测序以揭示FEV对巨噬细胞的分子调节。值得注意的是,我们发现FEV通过破坏线粒体来重新编程巨噬细胞的每一种代谢,并抑制氧化磷酸化和糖酵解。此外,FEV处理的巨噬细胞显示PPARγ表达降低,脂质处理表型改变,胆固醇流出减少,这可能会促进能源重新编程。一起来看,这些发现确定FEV是巨噬细胞重编程的驱动因素,提示通过口服FEV触发M2b巨噬细胞极化可能作为缓解CD中过度纤维化肠道疾病的策略。
    Faecalibacterium prausnitzii (F. prausnitzii) has been recognized for its various intestinal and extraintestinal benefits to human. And reduction of F. prausnitzii has been linked to an increased risk of intestinal fibrosis in patients of Crohn\'s disease (CD). In this study, oral administration of either live F. prausnitzii or its extracellular vesicles (FEVs) can markedly mitigate the severity of fibrosis in mice induced by repetitive administration of DSS. In vitro experiment revealed that FEVs were capable of directing the polarization of peripheral blood mononuclear cells (PBMCs) towards an M2b macrophage phenotype, which has been associated with anti-fibrotic activities. This effect of FEV was found to be stable under various conditions that promote the development of pro-fibrotic M1/M2a/M2c macrophages. Proteomics and RNA sequencing were performed to uncover the molecular modulation of macrophages by FEVs. Notably, we found that FEVs reprogramed every metabolism of macrophages by damaging the mitochondria, and inhibited oxidative phosphorylation and glycolysis. Moreover, FEV-treated macrophages showed a decreased expression of PPARγ and an altered lipid processing phenotype characterized by decreased cholesterol efflux, which may promote energy reprogramming. Taken together, these findings identify FEV as a driver of macrophage reprogramming, suggesting that triggering M2b macrophage polarization by oral admiration of FEV may serve as strategy to alleviate hyperfibrotic intestine conditions in CD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:肝脏是生酮的主要器官,而酮主要通过关键酶OXCT1在外周组织中代谢。我们以前发现,在肝细胞癌(HCC)细胞中,酮溶解通过OXCT1表达重新激活以促进肿瘤进展;然而,OXCT1是否调节抗肿瘤免疫仍不清楚.
    方法:探讨OXCT1在肝细胞癌中的体内表达规律,我们对人类HCC标本进行了多重免疫组织化学(mIHC)实验。探讨OXCT1在小鼠肝癌肿瘤相关巨噬细胞(TAMs)中的作用,我们产生了LysMcreOXCT1f/f(巨噬细胞中的OXCT1条件性敲除)小鼠。
    结果:这里,我们发现,抑制肿瘤相关巨噬细胞中OXCT1的表达通过琥珀酸-H3K4me3-Arg1轴减少CD8+T细胞耗竭.最初,我们发现OXCT1在稳态下在肝巨噬细胞中高表达,而OXCT在TAM中的表达进一步增加。巨噬细胞中的OXCT1缺乏通过将TAM重编程为抗肿瘤表型而抑制肿瘤生长,减少CD8+T细胞耗竭和增加CD8+T细胞的细胞毒性。机械上,高OXCT1表达诱导琥珀酸的积累,酮分解的副产品,在TAM中,它通过增加Arg1启动子中的H3K4三甲基化(H3K4me3)水平来促进Arg1转录。此外,匹莫齐特,OXCT1的抑制剂,抑制Arg1表达以及TAM向原瘤表型的极化,导致CD8+T细胞耗竭减少和肿瘤生长减速。最后,巨噬细胞中OXCT1的高表达与HCC患者的低生存率呈正相关.
    结论:结论:我们的结果表明,OXCT1表观遗传抑制抗肿瘤免疫,提示抑制TAMs中的OXCT1活性是治疗肝癌的有效方法。
    肝脏巨噬细胞的复杂代谢在塑造HCC进展和免疫调节中起着关键作用。靶向巨噬细胞代谢以抵消免疫抑制是HCC的有希望的途径。这里,我们发现生酮基因OXCT1在肿瘤相关巨噬细胞中高表达,并通过将TAMs重编程为原瘤表型促进肿瘤生长.TAMs中OXCT1的战略药物干预或基因下调可增强抗肿瘤免疫力,减缓肿瘤生长。我们的结果表明,抑制TAM中的OXCT1活性是治疗肝癌的有效方法。
    OBJECTIVE: The liver is the main organ of ketogenesis, while ketones are mainly metabolized in peripheral tissues via the critical enzyme 3-oxoacid CoA-transferase 1 (OXCT1). We previously found that ketolysis is reactivated in hepatocellular carcinoma (HCC) cells through OXCT1 expression to promote tumor progression; however, whether OXCT1 regulates antitumor immunity remains unclear.
    METHODS: To investigate the expression pattern of OXCT1 in HCC in vivo, we conducted multiplex immunohistochemistry experiments on human HCC specimens. To explore the role of OXCT1 in mouse HCC tumor-associated macrophages (TAMs), we generated LysMcreOXCT1f/f (OXCT1 conditional knockout in macrophages) mice.
    RESULTS: Here, we found that inhibiting OXCT1 expression in tumor-associated macrophages reduced CD8+ T-cell exhaustion through the succinate-H3K4me3-Arg1 axis. Initially, we found that OXCT1 was highly expressed in liver macrophages under steady state and that OXCT expression was further increased in TAMs. OXCT1 deficiency in macrophages suppressed tumor growth by reprogramming TAMs toward an antitumor phenotype, reducing CD8+ T-cell exhaustion and increasing CD8+ T-cell cytotoxicity. Mechanistically, high OXCT1 expression induced the accumulation of succinate, a byproduct of ketolysis, in TAMs, which promoted Arg1 transcription by increasing the H3K4me3 level in the Arg1 promoter. In addition, pimozide, an inhibitor of OXCT1, suppressed Arg1 expression as well as TAM polarization toward the protumor phenotype, leading to decreased CD8+ T-cell exhaustion and slower tumor growth. Finally, high expression of OXCT1 in macrophages was positively associated with poor survival in patients with HCC.
    CONCLUSIONS: In conclusion, our results demonstrate that OXCT1 epigenetically suppresses antitumor immunity, suggesting that suppressing OXCT1 activity in TAMs could be an effective approach for treating liver cancer.
    UNASSIGNED: The intricate metabolism of liver macrophages plays a critical role in shaping hepatocellular carcinoma progression and immune modulation. Targeting macrophage metabolism to counteract immune suppression presents a promising avenue for hepatocellular carcinoma treatment. Herein, we found that the ketogenesis gene OXCT1 was highly expressed in tumor-associated macrophages (TAMs) and promoted tumor growth by reprogramming TAMs toward a protumor phenotype. Pharmacological targeting or genetic downregulation of OXCT1 in TAMs enhances antitumor immunity and slows tumor growth. Our results suggest that suppressing OXCT1 activity in TAMs could be an effective approach for treating liver cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    失调的皮肤微生物群和受损的免疫应答是不愈合的糖尿病伤口的主要病因学因素。当前的抗菌策略无法协调免疫反应,并且不加选择地根除伤口部位的细菌。加剧微生物群的不平衡。从益生菌对微生物群的有益影响中汲取灵感,我们配制了含有植物乳杆菌和低聚果糖(LP/FOS@Gel)的活的微生态水凝胶,以重塑失调的皮肤微生物群并恢复受损的免疫反应,培养有利于最佳伤口愈合的环境。LP/FOS@Gel充当“唤起者”,巧妙地整合皮肤微生态,促进乳酸菌的增殖,Ralstonia,Muibaculum,芽孢杆菌,和Allobaculum,同时根除定植的致病菌。同时,LP/FOS@Gel持续产生乳酸以引起修复性巨噬细胞反应并阻碍核因子κB途径的激活,有效缓解炎症。作为智能微生态系统,LP/FOS@Gel在愈合过程中恢复皮肤的主权,并有效地协调宿主免疫系统和微生物之间的和谐对话,从而促进糖尿病感染性伤口的愈合。这些显著的属性使得LP/FOS@Gel对于实用的临床应用非常有利。本文受版权保护。保留所有权利。
    Dysregulated skin microbiota and compromised immune responses are the major etiological factors for non-healing diabetic wounds. Current antibacterial strategies fail to orchestrate immune responses and indiscriminately eradicate bacteria at the wound site, exacerbating the imbalance of microbiota. Drawing inspiration from the beneficial impacts that probiotics possess on microbiota, a living microecological hydrogel containing Lactobacillus plantarum and fructooligosaccharide (LP/FOS@Gel) is formulated to remodel dysregulated skin microbiota and reinstate compromised immune responses, cultivating a conducive environment for optimal wound healing. LP/FOS@Gel acts as an \"evocator,\" skillfully integrating the skin microecology, promoting the proliferation of Lactobacillus, Ralstonia, Muribaculum, Bacillus, and Allobaculum, while eradicating colonized pathogenic bacteria. Concurrently, LP/FOS@Gel continuously generates lactic acid to elicit a reparative macrophage response and impede the activation of the nuclear factor kappa-B pathway, effectively alleviating inflammation. As an intelligent microecological system, LP/FOS@Gel reinstates the skin\'s sovereignty during the healing process and effectively orchestrates the harmonious dialogue between the host immune system and microorganisms, thereby fostering the healing of diabetic infectious wounds. These remarkable attributes render LP/FOS@Gel highly advantageous for pragmatic clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    免疫原性细胞死亡(ICD)在触发肿瘤微环境(TME)中的抗肿瘤免疫反应中起着至关重要的作用。最近,相当多的注意力一直集中在铁性上,一种由细胞内铁诱导的ICD,已被证明可改变TME的免疫沙漠状态。然而,在以免疫沙漠为特征的癌症中,比如前列腺癌,诱导高水平铁中毒的策略仍然有限。辐射肿瘤细胞衍生微粒(RMPs)是放疗模拟物,已被证明可以激活cGAS-STING通路,诱导肿瘤细胞铁性凋亡,抑制M2巨噬细胞极化。RMP还可以充当具有生物相容性的试剂的载体。在本研究中,我们设计了一种治疗系统,其中将铁凋亡诱导物RSL-3加载到RMPs中,在使用RM-1细胞建立的体外和体内前列腺癌模型中进行了测试。凋亡诱导剂CT20肽(CT20p)也被添加到RMPs中以加重铁凋亡。我们的结果表明,RSL-3-和CT20p负载的RMPs(RC@RMPs)导致RM-1细胞的铁凋亡和凋亡。此外,CT20p通过促进活性氧(ROS)的产生对铁凋亡具有协同作用,脂质过氧化氢生产,和线粒体不稳定。RC@RMPs升高的树突状细胞(DC)表达MHCII,CD80和CD86促进M1巨噬细胞极化。在小鼠皮下移植的RM-1肿瘤模型中,RC@RMPs通过DC激活抑制肿瘤生长并延长生存时间,巨噬细胞重编程,增强CD8+T细胞浸润,和肿瘤中促炎细胞因子的产生。此外,抗PD-1联合治疗可改善RM-1肿瘤抑制。这项研究为前列腺癌免疫疗法的铁凋亡的协同增强提供了策略。
    Immunogenic cell death (ICD) plays a crucial role in triggering the antitumor immune response in the tumor microenvironment (TME). Recently, considerable attention has been dedicated to ferroptosis, a type of ICD that is induced by intracellular iron and has been demonstrated to change the immune desert status of the TME. However, among cancers that are characterized by an immune desert, such as prostate cancer, strategies for inducing high levels of ferroptosis remain limited. Radiated tumor cell-derived microparticles (RMPs) are radiotherapy mimetics that have been shown to activate the cGAS-STING pathway, induce tumor cell ferroptosis, and inhibit M2 macrophage polarization. RMPs can also act as carriers of agents with biocompatibility. In the present study, we designed a therapeutic system wherein the ferroptosis inducer RSL-3 was loaded into RMPs, which were tested in in vitro and in vivo prostate carcinoma models established using RM-1 cells. The apoptosis inducer CT20 peptide (CT20p) was also added to the RMPs to aggravate ferroptosis. Our results showed that RSL-3- and CT20p-loaded RMPs (RC@RMPs) led to ferroptosis and apoptosis of RM-1 cells. Moreover, CT20p had a synergistic effect on ferroptosis by promoting reactive oxygen species (ROS) production, lipid hydroperoxide production, and mitochondrial instability. RC@RMPs elevated dendritic cell (DC) expression of MHCII, CD80, and CD86 and facilitated M1 macrophage polarization. In a subcutaneously transplanted RM-1 tumor model in mice, RC@RMPs inhibited tumor growth and prolonged survival time via DC activation, macrophage reprogramming, enhancement of CD8+ T cell infiltration, and proinflammatory cytokine production in the tumor. Moreover, combination treatment with anti-PD-1 improved RM-1 tumor inhibition. This study provides a strategy for the synergistic enhancement of ferroptosis for prostate cancer immunotherapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    虽然磷酸钙已广泛用于骨科应用,如脊柱,四肢,牙科,和颌面外科,骨诱导性的缺乏往往阻碍了其治疗由病理微环境如肿瘤手术引起的骨缺损的有效性,骨质疏松,骨髓炎,和糖尿病。因此,本研究开发了一种基于镁掺杂生物活性玻璃的新型骨水泥。镁离子的适度释放通过控制羟基磷灰石的晶体尺寸来改善机械性能。通过对元素含量和热处理温度的详细讨论,发现2Mg-BG-800适用于骨水泥的构建。2Mg-BG-BC表现出良好的初始(15分钟)和最终(30分钟)凝固时间,抗压强度(29.45MPa),压缩模量(1851.49MPa),可注射性,和形状适应性。此外,Mg-BG-BC具有增强BMSCs成骨分化能力,并诱导巨噬细胞向M2表型极化,提示其骨质疏松性骨折再生的潜力。
    Although calcium phosphate has been extensively utilized in orthopedic applications such as spine, limbs, dentistry, and maxillofacial surgery, the lack of osteoinductive properties often hinders its effectiveness in treating bone defects resulting from pathological micro-environment such as tumor surgery, osteoporosis, osteomyelitis, and diabetic. Therefore, a novel bone cement based on magnesium-doped bioactive glass was developed in this study. The moderate release of magnesium ions improved the mechanical properties by controlling the crystal size of hydroxyapatite. Through detailed discussion of element content and heat treatment temperature, it was found that 2Mg-BG-800 was suitable for the construction of bone cement. 2Mg-BG-BC exhibited favorable initial (15 min) and final (30 min) setting time, compressive strength (29.45 MPa), compressive modulus (1851.49 MPa), injectability, and shape-adaptability. Furthermore, Mg-BG-BC demonstrated the ability to enhance the osteogenic differentiation of BMSCs, and induce macrophage polarization towards the M2 phenotype, suggesting its potential for osteoporotic fracture regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    通过Notch受体的信号内在地调节肿瘤细胞的发育和生长。这里,我们研究了Notch配体Jagged2在非小细胞肺癌(NSCLC)免疫逃避中的作用.在NSCLC中JAG2的高表达与生存呈负相关。在NSCLC临床前模型中,在癌细胞中Jag2而不是Jag1的缺失减弱了肿瘤生长并激活了保护性抗肿瘤T细胞应答。Jag2-/-肺肿瘤表现出更高频率的表达免疫刺激介质并触发T细胞依赖性抗肿瘤免疫的巨噬细胞。机械上,Jag2消融促进Nr4a介导的Notch配体DLL1/4对癌细胞的诱导。DLL1/4启动的Notch1/2信号在巨噬细胞中诱导转录因子IRF4的表达和巨噬细胞免疫刺激功能。IRF4表达是肺肿瘤中Jag2缺失的抗肿瘤作用所必需的。Jagged2的抗体靶向抑制肿瘤生长并激活IRF4驱动的巨噬细胞介导的抗肿瘤免疫。因此,Jagged2协调NSCLC中的免疫抑制系统,其可以被克服以激发巨噬细胞介导的抗肿瘤免疫。
    Signaling through Notch receptors intrinsically regulates tumor cell development and growth. Here, we studied the role of the Notch ligand Jagged2 on immune evasion in non-small cell lung cancer (NSCLC). Higher expression of JAG2 in NSCLC negatively correlated with survival. In NSCLC pre-clinical models, deletion of Jag2, but not Jag1, in cancer cells attenuated tumor growth and activated protective anti-tumor T cell responses. Jag2-/- lung tumors exhibited higher frequencies of macrophages that expressed immunostimulatory mediators and triggered T cell-dependent anti-tumor immunity. Mechanistically, Jag2 ablation promoted Nr4a-mediated induction of Notch ligands DLL1/4 on cancer cells. DLL1/4-initiated Notch1/2 signaling in macrophages induced the expression of transcription factor IRF4 and macrophage immunostimulatory functionality. IRF4 expression was required for the anti-tumor effects of Jag2 deletion in lung tumors. Antibody targeting of Jagged2 inhibited tumor growth and activated IRF4-driven macrophage-mediated anti-tumor immunity. Thus, Jagged2 orchestrates immunosuppressive systems in NSCLC that can be overcome to incite macrophage-mediated anti-tumor immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肝脏缺血再灌注损伤(HIRI)仍然是肝脏手术后器官衰竭的未解决的危险因素。我们的临床回顾性研究表明,较低的供体肝脏CX3-C趋化因子受体-1(CX3CR1)mRNA表达水平与上调的前分辨巨噬细胞受体MERTK相关,以及促进肝移植中同种异体移植损伤的恢复效率。为了进一步表征CX3CR1在调节HIRI中的作用,我们采用Wt和Cx3cr1-/-小鼠的小鼠肝脏部分热缺血再灌注模型,再灌注时间从6h延长至4-7天。Kupffer细胞(KCs)被clodronate脂质体(CL)提前耗尽,专注于浸润的巨噬细胞,再增殖动力学由FACS测定,IF和RNA-Seq.腹膜内注射CX3CR1拮抗剂AZD8797以询问潜在的药理学治疗策略。LXR激动剂DMHCA体外原代骨髓巨噬细胞(BMMs)培养,以及分子和功能研究,进行解剖CX3CR1在调节巨噬细胞细胞生物学发育和解决功能中的作用。我们观察到CX3CR1的缺乏或药理学抑制通过促进巨噬细胞以CCR1/CCR5方式迁移促进HIRI的解决,以及增强的MerTK介导的红细胞增多。我们的研究证明了CX3CR1在HIRI进展中的关键作用,并将其确定为临床肝移植的潜在治疗靶标。
    Hepatic ischemia-reperfusion injury(HIRI) remains to be an unsolved risk factor that contributes to organ failure after liver surgery. Our clinical retrospective study showed that lower donor liver CX3-C chemokine receptor-1(CX3CR1) mRNA expression level were correlated with upregulated pro-resolved macrophage receptor MERTK, as well as promoted restoration efficiency of allograft injury in liver transplant. To further characterize roles of CX3CR1 in regulating resolution of HIRI, we employed murine liver partial warm ischemia-reperfusion model by Wt & Cx3cr1-/- mice and the reperfusion time was prolonged from 6 h to 4-7 days. Kupffer cells(KCs) were depleted by clodronate liposome(CL) in advance to focus on infiltrating macrophages, and repopulation kinetics were determined by FACS, IF and RNA-Seq. CX3CR1 antagonist AZD8797 was injected i.p. to interrogate potential pharmacological therapeutic strategies. In vitro primary bone marrow macrophages(BMMs) culture by LXR agonist DMHCA, as well as molecular and functional studies, were undertaken to dissect roles of CX3CR1 in modulating macrophages cytobiological development and resolutive functions. We observed that deficiency or pharmacological inhibition of CX3CR1 facilitated HIRI resolution via promoted macrophages migration in CCR1/CCR5 manner, as well as enhanced MerTK-mediated efferocytosis. Our study demonstrated the critical roles of CX3CR1 in progression of HIRI and identified it as a potential therapeutic target in clinical liver transplantation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    单核细胞来源的巨噬细胞在炎症性疾病中起着关键的致病作用。在类风湿性关节炎(RA)的情况下,特定滑膜组织浸润巨噬细胞亚群的存在与活动性疾病或炎症消退有关.JAK抑制剂(JAKi)是第一个被批准用于治疗RA的靶向合成疾病缓解抗风湿药(tsDMARD),其疗效与生物制剂相当。然而,JAKi对巨噬细胞特化和分化的影响目前尚不清楚.我们分析了JAKi对RA患者外周血单核细胞亚群的转录和功能影响,以及对粒细胞-巨噬细胞集落刺激因子(GM-CSF)促进的单核细胞衍生巨噬细胞分化的影响,是推动RA发展和发病的因素。我们现在报道,JAKiUpadacitinib恢复了RA患者外周血单核细胞亚群的平衡,并以剂量依赖的方式使巨噬细胞偏向于获得抗炎转录和功能谱。Upadacitinib处理的巨噬细胞显示出定义与稳态/炎症消退相关的滑膜巨噬细胞的基因的强阳性富集。具体来说,Upadacitinib处理的巨噬细胞显示MAFB和MAFB调节基因的表达显着升高,GSK3β的抑制磷酸化升高,和更高的吞噬活性,并在病原刺激激活后显示出抗炎细胞因子谱。这些结果也由暴露于其他JAKI(baricitinib,托法替尼),但不存在TYK2抑制剂deucravitinib。作为一个整体,我们的结果表明,JAKi促进巨噬细胞重编程,以获得更多的抗炎/促分辨率谱,与JAKi增强MAFB表达能力相关的作用。
    Monocyte-derived macrophages play a key pathogenic role in inflammatory diseases. In the case of rheumatoid arthritis (RA), the presence of specific synovial tissue-infiltrating macrophage subsets is associated with either active disease or inflammation resolution. JAK inhibitors (JAKi) are the first targeted synthetic disease-modifying antirheumatic drugs (tsDMARD) approved for treatment of RA with comparable efficacy to biologics. However, the effects of JAKi on macrophage specification and differentiation are currently unknown. We have analyzed the transcriptional and functional effects of JAKi on human peripheral blood monocyte subsets from RA patients and on the differentiation of monocyte-derived macrophages promoted by granulocyte-macrophage colony-stimulating factor (GM-CSF), a factor that drives the development and pathogenesis of RA. We now report that JAKi Upadacitinib restores the balance of peripheral blood monocyte subsets in RA patients and skewed macrophages towards the acquisition of an anti-inflammatory transcriptional and functional profile in a dose-dependent manner. Upadacitinib-treated macrophages showed a strong positive enrichment of the genes that define synovial macrophages associated to homeostasis/inflammation resolution. Specifically, Upadacitinib-treated macrophages exhibited significantly elevated expression of MAFB and MAFB-regulated genes, elevated inhibitory phosphorylation of GSK3β, and higher phagocytic activity and showed an anti-inflammatory cytokine profile upon activation by pathogenic stimuli. These outcomes were also shared by macrophages exposed to other JAKi (baricitinib, tofacitinib), but not in the presence of the TYK2 inhibitor deucravacitinib. As a whole, our results indicate that JAKi promote macrophage re-programming towards the acquisition of a more anti-inflammatory/pro-resolution profile, an effect that correlates with the ability of JAKi to enhance MAFB expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    最近,使用纳米技术和复杂的递送方法靶向特定器官和细胞类型一直处于应用生物医学科学的最前沿。巨噬细胞是通过纳米递送进行免疫调节的有吸引力的靶标,因为它们大量涉及许多疾病的各个方面并且在其性质上是高度可塑性的。它们的功能“极化”状态的连续性多年来一直是研究的重点,从而对这些细胞的各个方面有了深刻的理解。单核细胞衍生的巨噬细胞从促炎到修复并因此产生促解决效应物的能力引起了人们对其治疗潜力的极大兴趣。这里,我们简要调查巨噬细胞个体发育和各种极化表型,突出它们在炎症分辨率转变中的功能。我们回顾了他们的诱导调解人,信号通路,和生物学程序,重点是核酸传感-IFN-I轴。我们还描绘了巨噬细胞的偏振光谱和它们在不同亚型之间的转变特征。最后,我们强调了目前针对巨噬细胞的不同药物递送方法,重点是纳米靶向,这可能会导致伤口愈合治疗的突破,骨再生,自身免疫,和纤维化疾病。
    Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional \"polarization\" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages\' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    特发性肺纤维化(IPF)的特征是纤维化基质沉积和不可逆的异常组织重塑。它们的作用机制与巨噬细胞的激活和受干扰的免疫环境有关。我们的目的是确定这些活化的巨噬细胞如何影响肺纤维化的发病机理。我们发现IPF患者的纤维化区域含有更多的血清和糖皮质激素诱导的激酶1(SGK1)阳性和M2型巨噬细胞。同样,博来霉素(BLM)+LPS显著触发IPF小鼠SGK1高表达,伴随着肺结构和功能的破坏,纤维化标志物增加和免疫微环境紊乱。机械上,SGK1通过触发糖原合酶激酶3β(GSK3β)-tat相互作用蛋白60(TIP60)-组蛋白3赖氨酸27乙酰化(H3K27ac)信号显着促进纤维化肺中M2型巨噬细胞的重编程,进一步释放趋化因子(C-C基序)配体9(CCL9)以吸引Th17细胞并将TGF-β递送至成纤维细胞以协同破坏免疫微环境,这在很大程度上被小鼠的巨噬细胞消耗所逆转。我们以巨噬细胞为切入点,深入分析IPF的发病机制,进一步为SGK1为代表的新药开发提供见解。
    Idiopathic pulmonary fibrosis (IPF) is characterized by fibrotic matrix deposition and irreversible aberrant tissue remodeling. Their mechanisms of action are associated with the activation of macrophages and a disturbed immune environment. We aim to determine how these activated macrophages influenced the pathogenesis of pulmonary fibrosis. We found the fibrotic areas of IPF patients contained more serum and glucocorticoid-induced kinase 1 (SGK1)-positive and M2-type macrophages. Similarly, bleomycin (BLM)+LPS significantly triggered high expression of SGK1 in the IPF mice, accompanied by destroyed lung structure and function, increased fibrosis markers and disturbed immune microenvironment. Mechanistically, SGK1 markedly promoted the reprogramming of M2-type macrophages in fibrotic lungs by triggering glycogen synthase kinase 3beta (GSK3β)-tat-interacting protein 60 (TIP60)- histone-3 lysine-27 acetylation (H3K27ac) signalings, which further released chemokine (C-C motif) ligand 9 (CCL9) to attract Th17 cells and delivered TGF-β to fibroblasts for synergistically destroying immune microenvironment, which was largely reversed by macrophage depletion in mice. We took macrophages as the entry point to deeply analyze IPF pathogenesis and further provided insights for the development of novel drugs represented by SGK1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号