Macrophages

巨噬细胞
  • 文章类型: Journal Article
    免疫细胞具有强烈的身体生活方式,其特征是结构可塑性和力量发挥。为了调查特定的免疫功能是否需要刻板的机械输出,我们使用超分辨率牵引力显微镜比较了细胞毒性T细胞形成的免疫突触与其他T细胞亚群和巨噬细胞形成的接触。T细胞突触是全局压缩的,这与与巨噬细胞吞噬相关的拉动和挤压有着根本的不同。每种细胞类型的力施加模式的光谱分解将细胞毒性与抗压强度联系起来,局部突出,和复杂的归纳,不对称地形。这些特征通过细胞骨架调节因子的遗传破坏被验证为细胞毒性驱动因子,突触分泌的实时成像,和界面变形的计算机模拟分析。突触结构和力的施加对目标刚度和尺寸敏感,这表明杀死的机械增强作用是生物物理适应性的。我们得出结论,细胞毒性和,言下之意,其他效应器响应由传出力的专门模式支持。
    Immune cells have intensely physical lifestyles characterized by structural plasticity and force exertion. To investigate whether specific immune functions require stereotyped mechanical outputs, we used super-resolution traction force microscopy to compare the immune synapses formed by cytotoxic T cells with contacts formed by other T cell subsets and by macrophages. T cell synapses were globally compressive, which was fundamentally different from the pulling and pinching associated with macrophage phagocytosis. Spectral decomposition of force exertion patterns from each cell type linked cytotoxicity to compressive strength, local protrusiveness, and the induction of complex, asymmetric topography. These features were validated as cytotoxic drivers by genetic disruption of cytoskeletal regulators, live imaging of synaptic secretion, and in silico analysis of interfacial distortion. Synapse architecture and force exertion were sensitive to target stiffness and size, suggesting that the mechanical potentiation of killing is biophysically adaptive. We conclude that cellular cytotoxicity and, by implication, other effector responses are supported by specialized patterns of efferent force.
    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
    溃疡性结肠炎(UC)的口服药物通常受到诸如积累不足等挑战的阻碍,粘液屏障的有限渗透,以及减轻过度ROS和炎性细胞因子的复杂任务。这里,我们提出了一种针对UC的靶向治疗的策略,该策略涉及海藻酸钠微球(SAMs),其中包含M2巨噬细胞膜(M2M)包被的Janus纳米马达(命名为Motor@M2M).SAM提供保护屏障,确保Motor@M2M能够承受恶劣的胃环境,并表现出受控的释放。M2M增强纳米马达对炎性组织的靶向精度并且充当炎性细胞因子的中和的诱饵。MnO2在氧化微环境中催化分解H2O2会产生O2气泡,推动马达@M2M穿过粘液屏障进入发炎的结肠组织。口服后,运动@M2M@SAM显著改善UC严重程度,包括炎症缓解,ROS清除,巨噬细胞重编程,以及肠道屏障和微生物群的恢复。因此,我们的研究介绍了一种有前途的口服微球配方的巨噬细胞-仿生纳米机器人,为UC治疗提供了一种有希望的方法。
    Oral medication for ulcerative colitis (UC) is often hindered by challenges such as inadequate accumulation, limited penetration of mucus barriers, and the intricate task of mitigating excessive ROS and inflammatory cytokines. Here, we present a strategy involving sodium alginate microspheres (SAMs) incorporating M2 macrophage membrane (M2M)-coated Janus nanomotors (denominated as Motor@M2M) for targeted treatment of UC. SAM provides a protective barrier, ensuring that Motor@M2M withstands the harsh gastric milieu and exhibits controlled release. M2M enhances the targeting precision of nanomotors to inflammatory tissues and acts as a decoy for the neutralization of inflammatory cytokines. Catalytic decomposition of H2O2 by MnO2 in the oxidative microenvironment generates O2 bubbles, propelling Motor@M2M across the mucus barrier into inflamed colon tissues. Upon oral administration, Motor@M2M@SAM notably ameliorated UC severity, including inflammation mitigation, ROS scavenging, macrophage reprogramming, and restoration of the intestinal barrier and microbiota. Consequently, our investigation introduces a promising oral microsphere formulation of macrophage-biomimetic nanorobots, providing a promising approach for UC treatment.
    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
    巨噬细胞在器官特异性功能和稳态中起关键作用。在肾上腺,巨噬细胞与产生醛固酮的肾小球带中的正弦毛细血管紧密相关。我们证明巨噬细胞保持毛细血管特化并调节醛固酮分泌。使用巨噬细胞特异性缺失VEGF-A,单细胞转录组学,和功能表型,我们发现VEGF-A的缺失会耗尽肾小球带中的PLVAP+有窗内皮细胞,导致基底膜胶原IV沉积增加和内皮下纤维化。这导致醛固酮分泌增加,称为“促分泌剂”信号。产生人醛固酮的腺瘤也显示毛细血管稀疏和基底膜增厚。骨髓细胞特异性VEGF-A缺失小鼠血清醛固酮升高,低钾血症,和高血压,模仿原发性醛固酮增多症。这些发现强调了巨噬细胞到内皮细胞的信号传导是内皮细胞特化所必需的,肾上腺功能,和血压调节,对其他内分泌器官有更广泛的影响。
    Macrophages play crucial roles in organ-specific functions and homeostasis. In the adrenal gland, macrophages closely associate with sinusoidal capillaries in the aldosterone-producing zona glomerulosa. We demonstrate that macrophages preserve capillary specialization and modulate aldosterone secretion. Using macrophage-specific deletion of VEGF-A, single-cell transcriptomics, and functional phenotyping, we found that the loss of VEGF-A depletes PLVAP+ fenestrated endothelial cells in the zona glomerulosa, leading to increased basement membrane collagen IV deposition and subendothelial fibrosis. This results in increased aldosterone secretion, called \"haptosecretagogue\" signaling. Human aldosterone-producing adenomas also show capillary rarefaction and basement membrane thickening. Mice with myeloid cell-specific VEGF-A deletion exhibit elevated serum aldosterone, hypokalemia, and hypertension, mimicking primary aldosteronism. These findings underscore macrophage-to-endothelial cell signaling as essential for endothelial cell specialization, adrenal gland function, and blood pressure regulation, with broader implications for other endocrine organs.
    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
    12,13-二羟基-9z-十八烯酸(12,13-DiHOME)是一种亚油酸二醇,衍生自细胞色素P-450(CYP)环氧合酶和环氧化物水解酶(EH)代谢。12,13-DiHOME与先天免疫反应中的炎症和线粒体损伤有关,但12,13-DiHOME如何促成这些影响尚不清楚。我们假设12,13-DiHOME通过影响NOD样受体蛋白3(NLRP3)炎性体激活来增强巨噬细胞炎症。为了检验这个假设,我们利用人单核细胞THP1细胞分化为巨噬细胞样细胞与佛波醇肉豆蔻酸盐乙酸(PMA)。在THP1巨噬细胞的脂多糖(LPS)引发过程中存在的12,13-DiHOME加剧了尼日利亚霉素诱导的NLRP3炎性体激活。使用高分辨率呼吸测量法,我们观察到用LPS+12,13-DiHOME引发改变线粒体呼吸功能。线粒体自噬,使用mito-Keima测量,也被引发期间存在的12,13-DiHOME调节。这些线粒体效应与LPS12,13-DiHOME引发的巨噬细胞对尼德霉素诱导的线粒体去极化和活性氧产生的敏感性增加有关。Ligericin诱导的线粒体损伤和NLRP3炎症小体激活的LPS12,13-DiHOME引发的巨噬细胞被线粒体钙单转体(MCU)抑制剂消融,鲁265.在LPS引发过程中存在的12,13-DiHOME也增强了在原代鼠骨髓来源的巨噬细胞中尼德霉素诱导的NLRP3炎性体激活。总之,这些数据表明12,13-DiHOME通过增强巨噬细胞中的NLRP3炎性体激活而具有促炎作用.
    12,13-dihydroxy-9z-octadecenoic acid (12,13-DiHOME) is a linoleic acid diol derived from cytochrome P-450 (CYP) epoxygenase and epoxide hydrolase (EH) metabolism. 12,13-DiHOME is associated with inflammation and mitochondrial damage in the innate immune response, but how 12,13-DiHOME contributes to these effects is unclear. We hypothesized that 12,13-DiHOME enhances macrophage inflammation through effects on NOD-like receptor protein 3 (NLRP3) inflammasome activation. To test this hypothesis, we utilized human monocytic THP1 cells differentiated into macrophage-like cells with phorbol myristate acetate (PMA). 12,13-DiHOME present during lipopolysaccharide (LPS)-priming of THP1 macrophages exacerbated nigericin-induced NLRP3 inflammasome activation. Using high-resolution respirometry, we observed that priming with LPS+12,13-DiHOME altered mitochondrial respiratory function. Mitophagy, measured using mito-Keima, was also modulated by 12,13-DiHOME present during priming. These mitochondrial effects were associated with increased sensitivity to nigericin-induced mitochondrial depolarization and reactive oxygen species production in LPS+12,13-DiHOME-primed macrophages. Nigericin-induced mitochondrial damage and NLRP3 inflammasome activation in LPS+12,13-DiHOME-primed macrophages were ablated by the mitochondrial calcium uniporter (MCU) inhibitor, Ru265. 12,13-DiHOME present during LPS-priming also enhanced nigericin-induced NLRP3 inflammasome activation in primary murine bone marrow-derived macrophages. In summary, these data demonstrate a pro-inflammatory role for 12,13-DiHOME by enhancing NLRP3 inflammasome activation in macrophages.
    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
    传统上,成功的疫苗依赖于特异性适应性免疫,通过用减毒的病原体激活淋巴细胞,或病原体亚基,在随后的暴露中引发更高的反应。然而,最近关于结核分枝杆菌和其他病原体的研究已经确定了“训练过的”单核细胞通过记忆样但非特异性免疫在保护中的作用。这里,我们使用体外共培养的方法来研究训练有素的巨噬细胞的潜在作用,包括肺泡巨噬细胞,在对土伦弗朗西斯菌活疫苗株(LVS)的免疫反应中。F.tularensis是一种在哺乳动物巨噬细胞内复制并引起呼吸道和全身性疾病的细胞内细菌。我们用F.tularensisLVS接种小鼠,然后获得肺泡巨噬细胞,或者来源于骨髓的巨噬细胞.LVS在两种类型的巨噬细胞中感染和复制,无论是幼稚的还是接种LVS的小鼠。然后将LVS感染的巨噬细胞与初始脾细胞共培养,皮内接种小鼠的脾细胞,或静脉接种小鼠的脾细胞。第一次,我们显示免疫(但不是幼稚)脾细胞控制肺泡巨噬细胞内的细菌复制,与以前使用骨髓源性巨噬细胞的结果相似。然而,在与未接种LVS疫苗的小鼠的初始巨噬细胞或巨噬细胞的共培养物之间,对营养内细菌复制的控制没有差异;此外,在所有条件下,上清液中的一氧化氮水平和干扰素-γ产量在很大程度上具有可比性.因此,在体外共培养的背景下,数据不支持皮内或静脉内接种LVS的小鼠的骨髓或肺中形成经过训练的巨噬细胞.
    目的:在单核细胞中发现非特异性的“训练免疫”产生了巨大的兴奋。然而,到目前为止,已经对相对较少的微生物进行了培训研究(例如,牛分枝杆菌卡介苗,用作疫苗的减毒活胞内细菌)和微生物物质(例如,LPS),目前尚不清楚感染期间的训练是否常见。我们以前证明了用土伦弗朗西斯菌活疫苗株(LVS)接种小鼠,另一种活的减毒细胞内细菌,免受无关细菌单核细胞增生李斯特菌的攻击。因此,本研究测试了LVS疫苗接种是否产生有助于这种保护的训练的巨噬细胞。要做到这一点,我们使用之前的体外共培养方法与鼠骨髓来源的巨噬细胞进行扩增和研究肺泡巨噬细胞.我们证明了肺泡巨噬细胞可以被生产性感染并用于表征与LVS免疫淋巴细胞的相互作用。然而,我们发现没有证据表明骨髓源性巨噬细胞或肺泡巨噬细胞接受了LVS疫苗的训练.
    Traditionally, successful vaccines rely on specific adaptive immunity by activating lymphocytes with an attenuated pathogen, or pathogen subunit, to elicit heightened responses upon subsequent exposures. However, recent work with Mycobacterium tuberculosis and other pathogens has identified a role for \"trained\" monocytes in protection through memory-like but non-specific immunity. Here, we used an in vitro co-culture approach to study the potential role of trained macrophages, including lung alveolar macrophages, in immune responses to the Live Vaccine Strain (LVS) of Francisella tularensis. F. tularensis is an intracellular bacterium that replicates within mammalian macrophages and causes respiratory as well as systemic disease. We vaccinated mice with F. tularensis LVS and then obtained lung alveolar macrophages, or derived macrophages from bone marrow. LVS infected and replicated comparably in both types of macrophages, whether naïve or from LVS-vaccinated mice. LVS-infected macrophages were then co-cultured with either naïve splenocytes, splenocytes from mice vaccinated intradermally, or splenocytes from mice vaccinated intravenously. For the first time, we show that immune (but not naïve) splenocytes controlled bacterial replication within alveolar macrophages, similar to previous results using bone marrow-derived macrophage. However, no differences in control of intramacrophage bacterial replication were found between co-cultures with naïve macrophages or macrophages from LVS-vaccinated mice; furthermore, nitric oxide levels and interferon-gamma production in supernatants were largely comparable across all conditions. Thus, in the context of in vitro co-cultures, the data do not support development of trained macrophages in bone marrow or lungs of mice vaccinated with LVS intradermally or intravenously.
    OBJECTIVE: The discovery of non-specific \"trained immunity\" in monocytes has generated substantial excitement. However, to date, training has been studied with relatively few microbes (e.g., Mycobacterium bovis Bacille Calmette-Guérin, a live attenuated intracellular bacterium used as a vaccine) and microbial substances (e.g., LPS), and it remains unclear whether training during infection is common. We previously demonstrated that vaccination of mice with Francisella tularensis Live Vaccine Strain (LVS), another live attenuated intracellular bacterium, protected against challenge with the unrelated bacterium Listeria monocytogenes. The present study therefore tested whether LVS vaccination engenders trained macrophages that contributed to this protection. To do so, we used a previous in vitro co-culture approach with murine bone marrow-derived macrophages to expand and study lung alveolar macrophages. We demonstrated that alveolar macrophages can be productively infected and employed to characterize interactions with LVS-immune lymphocytes. However, we find no evidence that either bone marrow-derived or alveolar macrophages are trained by LVS vaccination.
    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
    背景:风湿性心脏病(RHD)是由A组链球菌(GAS)感染引起的炎症细胞错误地攻击心脏瓣膜引起的,但目前尚不清楚哪些细胞或基因参与炎症细胞浸润瓣膜的过程。炎症浸润到靶组织需要增加磷酸化血管内皮钙黏着蛋白(p-VE-cad)的表达,p-VE-cad可增加内皮通透性,促进炎症细胞跨内皮迁移。P-VE-cad可能受RAS相关的C3肉毒杆菌底物1(RAC1)调节,连同磷酸化的富含脯氨酸的酪氨酸激酶2(p-PYK2)。而RAC1/p-PYK2/p-VE-cad是由血管细胞粘附分子-1(VCAM-1)的激活而触发的。VCAM-1与M1巨噬细胞通过极晚期抗原4(VLA4)粘附于内皮有关。炎症浸润进入瓣膜在RHD的早期发病机制中极为重要。然而,关于M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad是否参与RHD,尚无相关研究;因此,我们在这项研究中探索的是是否涉及M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad。
    方法:我们建立了大鼠RHD模型和M1巨噬细胞与内皮细胞共培养的细胞模型。随后,我们测量了炎症细胞浸润的程度,IL-6/IL-17水平,纤维化程度(COL3/1),RHD大鼠心脏瓣膜纤维化标志物(FSP1、COL1A1和COL3A1)的表达水平。此外,我们检测了M1/M2巨噬细胞生物标志物在大鼠模型和细胞模型中的表达,以及M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad的表达。我们还测试了M1巨噬细胞和内皮细胞共培养后内皮通透性的变化。
    结果:与对照组相比,RHD大鼠心脏瓣膜炎症细胞浸润和纤维化因子水平明显增高;M1巨噬细胞生物标志物(iNOS,CD86和TNF-α)在RHD大鼠中的表达显著高于M2巨噬细胞生物标志物(Arg1和TGF-β)。RHD年夜鼠心脏中VLA4/VCAM-1和RAC1/p-PYK2/p-VE-cad的表达程度显著增高。在细胞层面,M1巨噬细胞与内皮细胞共培养后,VLA4/VCAM-1和RAC1/p-PYK2/p-VE-cad的表达水平明显增高,由于与M1巨噬细胞共培养,内皮的通透性明显更大。
    结论:所有结果表明,M1巨噬细胞和VLA4/VCAM-1通路可能参与了RHD的炎症浸润过程。
    BACKGROUND: Rheumatic heart disease (RHD) is caused by inflammatory cells mistakenly attacking the heart valve due to Group A Streptococcus (GAS) infection, but it is still unclear which cells or genes are involved in the process of inflammatory cells infiltrating the valve. Inflammatory infiltration into the target tissue requires an increase in the expression of phosphorylated vascular endothelial-cadherin (p-VE-cad), p-VE-cad can increase the endothelial permeability and promote the migration of inflammatory cells across the endothelium. P-VE-cad is potentially regulated by RAS-related C3 botulinum substrate 1 (RAC1), together with phosphorylated proline-rich tyrosine kinase 2 (p-PYK2). While RAC1/p-PYK2/p-VE-cad is triggered by the activation of vascular cell adhesion molecule-1 (VCAM-1). VCAM-1 is related to M1 macrophages adhering to the endothelium via very late antigen 4 (VLA4). Inflammatory infiltration into the valve is extremely important in the early pathogenesis of RHD. However, there is no relevant research on whether M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad is involved in RHD; therefore, what we explored in this study was whether M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad is involved.
    METHODS: We established a rat model of RHD and a cell model of M1 macrophage and endothelial cell cocultivation. Subsequently, we measured the degree of inflammatory cell infiltration, the levels of IL-6/IL-17, the degree of fibrosis (COL3/1), and the expression levels of fibrosis markers (FSP1, COL1A1 and COL3A1) in the heart valves of RHD rats. Additionally, we detected the expression of M1/M2 macrophage biomarkers in rat model and cell model, as well as the expression of M1/VLA4/VCAM-1/RAC1/p-PYK2/p-VE-cad. We also tested the changes in endothelial permeability after coculturing M1 macrophages and endothelial cells.
    RESULTS: Compared to those in the control group, the levels of inflammatory cell infiltration and fibrotic factors in the heart valves of RHD rats were significantly higher; the expression of M1 macrophage biomarkers (iNOS, CD86 and TNF-α) in RHD rats was significantly higher; and significantly higher than the expression of M2 macrophage biomarkers (Arg1 and TGF-β). And the expression levels of VLA4/VCAM-1 and RAC1/p-PYK2/p-VE-cad in the hearts of RHD rats were significantly higher. At the cellular level, after coculturing M1 macrophages with endothelial cells, the expression levels of VLA4/VCAM-1 and RAC1/p-PYK2/p-VE-cad were significantly higher, and the permeability of the endothelium was significantly greater due to cocultivation with M1 macrophages.
    CONCLUSIONS: All the results suggested that M1 macrophages and the VLA4/VCAM-1 pathway are potentially involved in the process of inflammatory infiltration in RHD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    腹主动脉瘤(AAA)是一种威胁生命的疾病,直到其急剧破裂才被发现。由于缺乏有效的药物治疗,迫切需要探索新的预防和治疗策略。代谢重编程是细胞改变其代谢模式以满足物质和能量需求的细胞过程,包括葡萄糖代谢,脂质代谢和氨基酸代谢。最近,代谢重编程在心血管疾病中的调节作用,尤其是AAA,引起了极大的关注。本文就血管平滑肌细胞(VSMCs)和巨噬细胞代谢重编程对AAA发生发展影响的研究进展作一综述。特别是它们在VSMCs凋亡和表型转化等主要病理过程中的作用,细胞外基质重塑,氧化应激,和炎症反应。旨在从代谢的角度为AAA的机制研究和临床治疗提供新的线索。
    Abdominal aortic aneurysm (AAA) is a life-threatening disease that remains undetected until it acutely ruptures. Due to lack of effective pharmaceutic therapies, it is urgent to explore new prevention and treatment strategies. Metabolic reprogramming is a cellular process through which cells change their metabolic patterns to meet material and energy requirements, including glucose metabolism, lipid metabolism and amino acid metabolism. Recently, the regulatory role of metabolic reprogramming in cardiovascular diseases, especially AAA, has attracted significant attention. This review article focuses on the research progress regarding the effects of metabolic reprogramming of vascular smooth muscle cells (VSMCs) and macrophages on the occurrence and development of AAA, especially their roles in major pathological processes such as VSMCs apoptosis and phenotype transformation, extracellular matrix remodeling, oxidative stress, and inflammatory response. The aim is to provide new clues for the mechanism research and clinical treatment of AAA from the perspective of metabolism.
    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
    最近的研究表明,透明质酸(HA)对牙周临床结果具有积极作用。这项体外研究旨在研究四种不同的HA对牙周生物膜和免疫细胞之间相互作用的影响。
    包括四种HA:高分子量HA(HHA,非交联),低分子量HA(LHA),低聚物HA(OHA),和交联的高分子量HA(CHA)。进行了一系列实验以验证HA对以下方面的影响:(i)12种牙周生物膜(形成和预先存在);(ii)在暴露或不暴露于牙周生物膜的情况下,单核细胞(MONO-MAC-6)细胞和牙周膜成纤维细胞(PDLF)中炎性细胞因子和HA受体的表达;(iii)在MONO-MAC-6细胞和PDLF中产生活性氧(ROS),并存在生物膜和HA
    结果表明,HHA和CHA减少了新形成的(4小时)生物膜和预先存在的五天大生物膜中的细菌数量。没有生物膜的挑战,OHA通过增加MONO-MAC细胞中IL-1β和IL-10水平和PDLF中IL-8水平以时间依赖性方式引发炎症反应,而CHA通过抑制MONO-MAC细胞中IL-10和PDLF中IL-8的表达来抑制这种反应。在生物膜的挑战下,HA以分子量依赖性方式降低了IL-1β的表达(最多降低了HHA),并增加了MONO-MAC-6细胞中的IL-10水平(最多增加了CHA)。HA和两种细胞之间的相互作用可以通过ICAM-1受体发生。生物膜刺激增加MONO-MAC-6细胞和PDLF中的ROS水平,但只有HHA轻微抑制了两个细胞中由生物膜刺激诱导的ROS的高生成。
    总的来说,这些结果表明OHA诱导炎症,而HHA和CHA表现出抗生物膜,主要是抗炎,和牙周环境中的抗氧化特性。
    UNASSIGNED: Recent studies have demonstrated a positive role of hyaluronic acid (HA) on periodontal clinical outcomes. This in-vitro study aimed to investigate the impact of four different HAs on interactions between periodontal biofilm and immune cells.
    UNASSIGNED: The four HAs included: high-molecular-weight HA (HHA, non-cross-linked), low-molecular-weight HA (LHA), oligomers HA (OHA), and cross-linked high-molecular-weight HA (CHA). Serial experiments were conducted to verify the influence of HAs on: (i) 12-species periodontal biofilm (formation and pre-existing); (ii) expression of inflammatory cytokines and HA receptors in monocytic (MONO-MAC-6) cells and periodontal ligament fibroblasts (PDLF) with or without exposure to periodontal biofilms; (iii) generation of reactive oxygen species (ROS) in MONO-MAC-6 cells and PDLF with presence of biofilm and HA.
    UNASSIGNED: The results indicated that HHA and CHA reduced the bacterial counts in a newly formed (4-h) biofilm and in a pre-existing five-day-old biofilm. Without biofilm challenge, OHA triggered inflammatory reaction by increasing IL-1β and IL-10 levels in MONO-MAC cells and IL-8 in PDLF in a time-dependent manner, whereas CHA suppressed this response by inhibiting the expression of IL-10 in MONO-MAC cells and IL-8 in PDLF. Under biofilm challenge, HA decreased the expression of IL-1β (most decreasing HHA) and increased IL-10 levels in MONO-MAC-6 cells in a molecular weight dependent manner (most increasing CHA). The interaction between HA and both cells may occur via ICAM-1 receptor. Biofilm stimulus increased ROS levels in MONO-MAC-6 cells and PDLF, but only HHA slightly suppressed the high generation of ROS induced by biofilm stimulation in both cells.
    UNASSIGNED: Overall, these results indicate that OHA induces inflammation, while HHA and CHA exhibit anti-biofilm, primarily anti-inflammatory, and antioxidant properties in the periodontal environment.
    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
    生物钟的破坏与炎症和免疫疾病有关。BMAL2,一种关键的昼夜节律蛋白,与时钟形成二聚体,激活转录。细胞外冷诱导RNA结合蛋白(eCIRP),在脓毒症期间释放,能诱导巨噬细胞内毒素耐受。我们假设eCIRP通过触发在骨髓细胞上表达的受体-1(TREM-1)诱导BMAL2表达并促进巨噬细胞内毒素耐受。
    C57BL/6野生型(WT)雄性小鼠通过盲肠结扎穿孔(CLP)进行脓毒症。通过ELISA评估CLP后20小时的eCIRP血清水平。用各种剂量的重组小鼠(rm)CIRP(eCIRP)处理腹膜巨噬细胞(PerM)24小时。然后用LPS刺激细胞5小时。通过ELISA评估培养上清液中TNF-α和IL-6的水平。PerM用eCIRP处理24小时,并通过qPCR评估PD-L1,IL-10,STAT3,TREM-1和昼夜节律基因如BMAL2,CRY1和PER2的表达。使用来自TREM-1-/-小鼠的PerM通过qPCR测定TREM-1对eCIRP诱导的PerM内毒素耐受性和PD-L1、IL-10和STAT3表达的影响。通过PCR阵列确定eCIRP处理的巨噬细胞中的昼夜节律基因表达谱,并通过qPCR确认。通过转染BMAL2CRISPR活化质粒在骨髓来源的巨噬细胞中进行BMAL2活化的诱导。通过计算建模确定BMAL2在PD-L1启动子中的相互作用,并通过BIAcore测定进行确认。
    与假手术小鼠相比,在败血症小鼠中eCIRP的血清水平增加。用eCIRP预处理的巨噬细胞在LPS攻击后显示TNFα和IL-6释放减少,表明巨噬细胞内毒素耐受。此外,eCIRP增加免疫耐受标志物PD-L1、IL-10和STAT3的表达。有趣的是,TREM-1缺乏可逆转eCIRP诱导的巨噬细胞内毒素耐受,并显著降低PD-L1、IL-10和STAT3表达。用eCIRP处理的腹膜巨噬细胞中昼夜节律基因的PCR阵列筛选显示BMAL2,CRY1和PER2的表达升高。在eCIRP处理的巨噬细胞中,TREM-1缺乏阻止了这些昼夜节律基因的上调。在巨噬细胞中,可诱导的BMAL2表达与PD-L1表达增加相关。在败血症的人类患者中,与健康受试者相比,血液单核细胞显示BMAL2和PD-L1的表达增加.计算建模和BIAcore测定确定了PD-L1启动子中BMAL2的推定结合区,提示BMAL2正调节巨噬细胞中PD-L1的表达。
    eCIRP通过TREM-1上调BMAL2的表达,导致脓毒症中巨噬细胞内毒素耐受。靶向eCIRP以维持昼夜节律可以纠正内毒素耐受性并增强宿主对细菌感染的抵抗力。
    UNASSIGNED: The disruption of the circadian clock is associated with inflammatory and immunological disorders. BMAL2, a critical circadian protein, forms a dimer with CLOCK, activating transcription. Extracellular cold-inducible RNA-binding protein (eCIRP), released during sepsis, can induce macrophage endotoxin tolerance. We hypothesized that eCIRP induces BMAL2 expression and promotes macrophage endotoxin tolerance through triggering receptor expressed on myeloid cells-1 (TREM-1).
    UNASSIGNED: C57BL/6 wild-type (WT) male mice were subjected to sepsis by cecal ligation and puncture (CLP). Serum levels of eCIRP 20 h post-CLP were assessed by ELISA. Peritoneal macrophages (PerM) were treated with recombinant mouse (rm) CIRP (eCIRP) at various doses for 24 h. The cells were then stimulated with LPS for 5 h. The levels of TNF-α and IL-6 in the culture supernatants were assessed by ELISA. PerM were treated with eCIRP for 24 h, and the expression of PD-L1, IL-10, STAT3, TREM-1 and circadian genes such as BMAL2, CRY1, and PER2 was assessed by qPCR. Effect of TREM-1 on eCIRP-induced PerM endotoxin tolerance and PD-L1, IL-10, and STAT3 expression was determined by qPCR using PerM from TREM-1-/- mice. Circadian gene expression profiles in eCIRP-treated macrophages were determined by PCR array and confirmed by qPCR. Induction of BMAL2 activation in bone marrow-derived macrophages was performed by transfection of BMAL2 CRISPR activation plasmid. The interaction of BMAL2 in the PD-L1 promoter was determined by computational modeling and confirmed by the BIAcore assay.
    UNASSIGNED: Serum levels of eCIRP were increased in septic mice compared to sham mice. Macrophages pre-treated with eCIRP exhibited reduced TNFα and IL-6 release upon LPS challenge, indicating macrophage endotoxin tolerance. Additionally, eCIRP increased the expression of PD-L1, IL-10, and STAT3, markers of immune tolerance. Interestingly, TREM-1 deficiency reversed eCIRP-induced macrophage endotoxin tolerance and significantly decreased PD-L1, IL-10, and STAT3 expression. PCR array screening of circadian clock genes in peritoneal macrophages treated with eCIRP revealed the elevated expression of BMAL2, CRY1, and PER2. In eCIRP-treated macrophages, TREM-1 deficiency prevented the upregulation of these circadian genes. In macrophages, inducible BMAL2 expression correlated with increased PD-L1 expression. In septic human patients, blood monocytes exhibited increased expression of BMAL2 and PD-L1 in comparison to healthy subjects. Computational modeling and BIAcore assay identified a putative binding region of BMAL2 in the PD-L1 promoter, suggesting BMAL2 positively regulates PD-L1 expression in macrophages.
    UNASSIGNED: eCIRP upregulates BMAL2 expression via TREM-1, leading to macrophage endotoxin tolerance in sepsis. Targeting eCIRP to maintain circadian rhythm may correct endotoxin tolerance and enhance host resistance to bacterial infection.
    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
    越来越多的证据表明细胞外囊泡(EV)因子是细胞治疗的介质。心球来源的细胞是具有组织修复能力的心脏来源的细胞。wnt/β-连环蛋白信号的下游靶标的激活,色氨酸2,3双加氧酶(TDO2)使治疗惰性皮肤成纤维细胞具有心脏保护作用。这里,我们研究了来自TDO2增强成纤维细胞的浓缩条件培养基(TDO2-CCM)发挥心脏保护作用的机制。与来自常规成纤维细胞的CCM(HDF-CCM)相比,TDO2-CCM在MI的小鼠模型中具有心脏保护性。在24小时时心脏组织的转录组学分析证明,与HDF-CCM或载体相比,在给予TDO2-CCM的动物中广泛抑制炎症和细胞应激标志物。TDO2-EVRNA的测序分析表明,有大量的Y衍生的小RNA,称为“NT4”。通过尺寸排阻色谱和RNA酶保护测定纯化TDO2-EV表明NT4被封装在EV内部。与TDO2-CCM一致,暴露于NT4的巨噬细胞显示抑制炎症和细胞应激介质,特别是p21/cdkn1a。NT4耗尽的TDO2-CCM导致免疫调节能力降低。最后,在急性心肌梗死模型中,单独给予NT4具有心肌保护作用.一起来看,这些发现阐明了TDO2增强通过富集NT4介导分泌型EV效力的机制,NT4抑制了包括p21/cdkn1a在内的上游细胞应激介质.
    Mounting evidence implicates extracellular vesicles (EVs) factors as mediators of cell therapy. Cardiosphere-derived cells are cardiac-derived cells with tissue reparative capacity. Activation of a downstream target of wnt/β-catenin signalling, tryptophan 2,3 dioxygenase (TDO2) renders therapeutically inert skin fibroblasts cardioprotective. Here, we investigate the mechanism by which concentrated conditioned media from TDO2-augmented fibroblasts (TDO2-CCM) exert cardioprotective effects. TDO2-CCM is cardioprotective in a mouse model of MI compared to CCM from regular fibroblasts (HDF-CCM). Transcriptomic analysis of cardiac tissue at 24 h demonstrates broad suppression of inflammatory and cell stress markers in animals given TDO2-CCM compared to HDF-CCM or vehicle. Sequencing analysis of TDO2-EV RNA demonstrated abundance of a small Y-derived small RNA dubbed \'NT4\'. Purification of TDO2-EVs by size-exclusion chromatography and RNAse protection assays demonstrated that NT4 is encapsulated inside EVs. Consistently with TDO2-CCM, macrophages exposed to NT4 showed suppression of the inflammatory and cell stress mediators, particularly p21/cdkn1a. NT4-depleted TDO2-CCM resulted in diminished immunomodulatory capacity. Finally, administration of NT4 alone was cardioprotective in an acute model of myocardial infarction. Taken together, these findings elucidate the mechanism by which TDO2 augmentation mediates potency in secreted EVs through enrichment of NT4 which suppresses upstream cell stress mediators including p21/cdkn1a.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号