MyD88

MyD88
  • 文章类型: Journal Article
    简介:细胞分离技术和高端组学技术的最新进展允许研究多个组学领域的单细胞集,并彻底探索细胞功能和各种功能阶段。虽然大多数的研究集中在单细胞群体的双重RNA和蛋白质分析,包括脂质和代谢物谱分析以全面阐明控制细胞功能的分子机制和途径至关重要,以及不同功能阶段的表型。方法:为了解决这个差距,采用同时提取脂质的细胞脂质组学和转录组学表型方法,代谢物,和来自单细胞群体的RNA结合非靶向细胞4维(4D)-脂质组学分析以及RNA测序被开发为能够从最低可能数量的细胞进行全面的多组分子谱分析。利用参考细胞模型来确定这种多组学分析所需的最小细胞数。为了证明高分辨率细胞多组学在病理和生理条件下早期鉴定细胞表型变化的可行性,我们在两个不同激活阶段实施了这种方法对巨噬细胞进行表型分析:MyD88敲除巨噬细胞作为动脉粥样硬化保护的细胞模型。和野生型巨噬细胞。结果和讨论:这项多组学研究能够确定MyD88-KO获得的具有抗炎和动脉粥样硬化保护功能的巨噬细胞中的脂质含量重塑,因此加快了对免疫细胞效应子功能背后的分子机制和治疗干预可能的分子靶标的理解.这里显示磷脂酰胆碱和纤溶酶原/纤溶酶原的丰富功能作用伴随着巨噬细胞获得抗炎功能的遗传变化,这些发现可以作为巨噬细胞重编程研究以及对疾病的一般免疫和炎症反应的参考。
    Introduction: Recent progress in cell isolation technologies and high-end omic technologies has allowed investigation of single cell sets across multiple omic domains and a thorough exploration of cellular function and various functional stages. While most multi-omic studies focused on dual RNA and protein analysis of single cell population, it is crucial to include lipid and metabolite profiling to comprehensively elucidate molecular mechanisms and pathways governing cell function, as well as phenotype at different functional stages. Methods: To address this gap, a cellular lipidomics and transcriptomics phenotyping approach employing simultaneous extraction of lipids, metabolites, and RNA from single cell populations combined with untargeted cellular 4 dimensional (4D)-lipidomics profiling along with RNA sequencing was developed to enable comprehensive multi-omic molecular profiling from the lowest possible number of cells. Reference cell models were utilized to determine the minimum number of cells required for this multi-omics analysis. To demonstrate the feasibility of higher resolution cellular multi-omics in early-stage identification of cellular phenotype changes in pathological and physiological conditions we implemented this approach for phenotyping of macrophages in two different activation stages: MyD88-knockout macrophages as a cellular model for atherosclerosis protection, and wild type macrophages. Results and Discussion: This multi-omic study enabled the determination of the lipid content remodeling in macrophages with anti-inflammatory and atherosclerotic protective function acquired by MyD88-KO, hence expedites the understanding of the molecular mechanisms behind immune cells effector functionality and of possible molecular targets for therapeutic intervention. An enriched functional role of phosphatidylcholine and plasmenyl/plasmalogens was shown here to accompany genetic changes underlying macrophages acquisition of anti-inflammatory function, finding that can serve as reference for macrophages reprogramming studies and for general immune and inflammation response to diseases.
    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
    哺乳期乳腺炎是产后动物的一种使人衰弱的炎性乳腺疾病。髓样分化初级反应蛋白MyD88是先天模式识别受体toll样受体4(TLR4)的关键下游接头,在炎症中起重要作用。然而,乳腺上皮细胞中MyD88在乳腺炎进展中的具体作用尚未得到研究.在这项研究中,采用脂多糖(LPS)诱导的小鼠乳腺炎模型,细胞因子如Tnf-α,IL-1β,如通过实时qPCR所显示的,IL-6、Cxcl1、Cxcl2和Ccl2在炎性乳腺中显著增加。然而,在乳腺上皮细胞(cKO)中使用MyD88缺陷的小鼠显示Tnf-α的表达减少,IL-1β,与对照小鼠相比,乳腺中的IL-6,Cxcl1和Cxcl2,当受到LPS诱导的乳腺炎。裂解的caspase-3的免疫组织化学染色显示,在MyD88cKO小鼠中,炎症诱导的细胞凋亡减少。此外,MyD88cKO小鼠肺泡腔内浸润的炎症细胞明显减少,包括Ly6G阳性中性粒细胞和F4/80阳性巨噬细胞。在LPS处理的乳腺中的RNA-seq显示,与对照小鼠相比,MyD88cKO小鼠具有显著下调的炎症相关基因和上调的与抗炎过程和脂质代谢相关的基因。因此,这些结果表明,乳腺上皮细胞中的MyD88对于乳腺炎的进展至关重要。这项研究不仅对理解乳腺上皮细胞的先天免疫反应具有重要意义。但也可能有助于开发新的治疗乳腺炎的治疗药物。
    Lactation mastitis is a debilitating inflammatory mammary disease in postpartum animals. Myeloid differentiation primary response protein MyD88 is the key downstream adapter for innate pattern recognition receptor toll-like receptor 4 (TLR4), which plays an important role in inflammation. However, the specific role of MyD88 in mammary epithelial cells in the progression of mastitis has not been investigated. In this study, lipopolysaccharide (LPS)-induced mouse mastitis model was used and cytokines such as Tnf-α, Il-1β, Il-6, Cxcl1, Cxcl2 and Ccl2 were significantly increased in inflammatory mammary gland as shown by real time-qPCR. However, the mice with MyD88-deficienet in mammary epithelial cells (cKO) showed a reduction in the expression of Tnf-α, Il-1β, Il-6, Cxcl1 and Cxcl2 in mammary gland compared with control mice, when subjected to LPS induced mastitis. Immunohistochemical staining of cleaved caspase-3 showed that the cell apoptosis induced by inflammation were decreased in MyD88 cKO mice. Furthermore, there were significantly fewer infiltrating inflammatory cells in alveolar lumen of MyD88 cKO mice, including Ly6G-positive neutrophils and F4/80-positive macrophages. RNA-seq in LPS treated mammary glands showed that MyD88 cKO mice had significantly downregulated inflammation-related genes and upregulated genes related to anti-inflammation processes and lipid metabolism compared with control mice. Thus, these results demonstrate that MyD88 in mammary epithelial cells is essential for mastitis progression. And this study not only has important implications for understanding the innate immune response in mammary epithelial cells, but also potentially helps the development of new therapeutic drugs for treating mastitis.
    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
    包封在脂质纳米颗粒(LNP)中的mRNA治疗剂提供了用于治疗各种疾病的有希望的途径。虽然mRNA疫苗预期免疫原性,mRNA负载LNP的相关反应原性提出了重大挑战,特别是在需要多次给药的蛋白质替代疗法中,导致不良反应和次优的治疗结果。历史上,研究主要集中在mRNA货物的反应原性,在这种情况下,LNP的作用被低估了。LNP的调节和促炎特性,至少部分源于可电离的脂质,可能诱发炎症,激活toll样受体(TLRs),并影响mRNA翻译。在理解LNP诱导的TLR激活及其对动物疾病行为诱导的影响方面仍然存在知识差距。我们假设LNP中的可电离脂质,结构上类似于脂多糖的脂质A,可以通过MyD88和TRIF适配器激活TLR4信号,从而传播LNP相关的反应原性。我们利用基因消融研究和药理学受体操作进行的全面调查证明,LNP激活的TLR4在小鼠中触发了不同的生理上有意义的反应。我们表明TLR4和MyD88对于反应原信号启动是必不可少的,促炎基因表达,和生理结果,如食物摄入量和体重--小鼠疾病行为的稳健指标。TLR4抑制剂TAK-242的应用通过减轻TLR4驱动的炎症反应而有效地降低了与LNP相关的反应原性。我们的发现阐明了TLR4-MyD88轴在LNP诱导的反应原性中的关键作用,为开发更安全的mRNA疗法提供了机制框架,并提供了通过靶向抑制该途径来减轻不良反应的策略。
    mRNA therapeutics encapsulated in lipid nanoparticles (LNPs) offer promising avenues for treating various diseases. While mRNA vaccines anticipate immunogenicity, the associated reactogenicity of mRNA-loaded LNPs poses significant challenges, especially in protein replacement therapies requiring multiple administrations, leading to adverse effects and suboptimal therapeutic outcomes. Historically, research has primarily focused on the reactogenicity of mRNA cargo, leaving the role of LNPs understudied in this context. Adjuvanticity and pro-inflammatory characteristics of LNPs, originating at least in part from ionizable lipids, may induce inflammation, activate toll-like receptors (TLRs), and impact mRNA translation. Knowledge gaps remain in understanding LNP-induced TLR activation and its impact on induction of animal sickness behavior. We hypothesized that ionizable lipids in LNPs, structurally resembling lipid A from lipopolysaccharide, could activate TLR4 signaling via MyD88 and TRIF adaptors, thereby propagating LNP-associated reactogenicity. Our comprehensive investigation utilizing gene ablation studies and pharmacological receptor manipulation proves that TLR4 activation by LNPs triggers distinct physiologically meaningful responses in mice. We show that TLR4 and MyD88 are essential for reactogenic signal initiation, pro-inflammatory gene expression, and physiological outcomes like food intake and body weight─robust metrics of sickness behavior in mice. The application of the TLR4 inhibitor TAK-242 effectively reduces the reactogenicity associated with LNPs by mitigating TLR4-driven inflammatory responses. Our findings elucidate the critical role of the TLR4-MyD88 axis in LNP-induced reactogenicity, providing a mechanistic framework for developing safer mRNA therapeutics and offering a strategy to mitigate adverse effects through targeted inhibition of this pathway.
    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:他汀类药物是具有抗炎作用的降脂药。围绕他汀类药物在2019年冠状病毒病患者(新冠肺炎)中的益处的数据是相互矛盾的。我们试图更好地了解他汀类药物在新冠肺炎相关炎症中的影响。
    方法:我们利用了Covid-19炎症的国际研究,这是2020年2月至2022年10月期间因Covid-19住院的前瞻性多中心队列患者。参与者接受了炎症生物标志物的系统评估。我们使用逻辑回归模型和逆概率加权(IPTW)来检查先前使用他汀类药物与住院死亡的复合结局之间的关联。需要机械通风,需要肾脏替代治疗.
    结果:本研究共纳入4,464例患者,其中1,364人(27.5%)在入院前服用他汀类药物。有1,061个主要结局事件,包括540人死亡,854机械通气和313肾脏替代治疗。在炎症的生物标志物中,调整已知的混杂因素后,他汀类药物的使用仅与较低水平的可溶性尿激酶纤溶酶原激活物受体(suPAR)相关。在多变量逻辑回归分析中,与未使用他汀类药物的患者相比,使用他汀类药物的复合结局几率较低(校正比值比(aOR)0.63,95CI[0.53-0.76])。结果与IPTW(aOR0.92,95CI[0.89-0.95])一致。他汀类药物对suPAR介导的主要结果的影响比例估计为31.5%。
    结论:在因Covid-19住院的患者中,先前使用他汀类药物与改善预后和降低炎症相关。
    BACKGROUND: Statins are lipid-lowering agents with with anti-inflammatory effects. Data surrounding the benefits of statins in patients with coronavirus disease 2019 (Covid-19) are conflicting. We sought to better understand the impact of statins in the context of Covid-19-related inflammation.
    METHODS: We leveraged the International Study of Inflammation in Covid-19, a prospective multicenter cohort of patients hospitalized for Covid-19 between February 2020 and October 2022. Participants underwent systematic assessment of biomarkers of inflammation. We used logistic regression modeling and inverse probability-of-treatment weighting (IPTW) to examine the association between prior statin use and the composite outcome of in-hospital death, need for mechanical ventilation, and need for renal replacement therapy.
    RESULTS: A total of 4,464 patients were included in the study, of whom 1,364 (27.5%) were taking a statin prior to admission. There were 1,061 primary outcome events, including 540 deaths, 854 mechanical ventilation and 313 renal replacement therapy. Amongst biomarkers of inflammation, statin use was associated solely with lower levels of soluble urokinase plasminogen activator receptor (suPAR) after adjusting for known confounders. In multivariable logistic regression analysis, statin use was associated with lower odds of the composite outcome (adjusted odds ratio (aOR) 0.63, 95%CI[0.53-0.76]) compared to patients not on statins. Findings were consistent with IPTW (aOR 0.92, 95%CI [0.89- 0.95]). The proportion of the effect of statin on the primary outcome mediated by suPAR was estimated at 31.5%.
    CONCLUSIONS: Prior-statin use is associated with improved outcomes and lower inflammation as measured by suPAR levels in patients hospitalized for Covid-19.
    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
    暂无摘要。
    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
    全球有近600万人死于由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)感染引起的冠状病毒病(COVID-19)爆发。尽管COVID-19疫苗在很大程度上成功地降低了疾病的严重程度和死亡人数,疫苗诱导的免疫力随着时间的推移而下降,以及新的病毒变异体或突变的持续出现,突显了开发针对SARS-CoV-2的广谱宿主介导疗法的替代策略的必要性.严重COVID-19的一个关键特征是先天免疫信号失调,最终导致大量促炎细胞因子和趋化因子的高表达和缺乏抗病毒干扰素(IFN),特别是I型(α和β)和III型(λ)。作为自然宿主防御,髓样分化初级反应蛋白,MyD88通过Toll样受体(TLRs)的信号转导途径在先天和获得性免疫反应中起关键作用,一种病原体识别受体(PRR)。然而,最近的研究表明,病毒感染会上调MyD88的表达,并通过负调节I型IFN来损害宿主的抗病毒反应。半乳糖凝集素-3(Gal3),病毒感染的另一个关键角色,已显示通过调节病毒进入和激活TLRs来调节宿主免疫应答,NLRP3炎性体,和NF-κB,导致促炎细胞因子的释放,并有助于整体炎症反应,所谓的“细胞因子风暴”。这些研究表明,对MyD88和Gal3的特异性抑制可能是一种有希望的COVID-19治疗方法。这篇综述提出了MyD88和Gal3靶向抗病毒药物发现的未来方向,强调在SARS-CoV-2感染中恢复宿主免疫力的潜力。
    Nearly six million people worldwide have died from the coronavirus disease (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although COVID-19 vaccines are largely successful in reducing the severity of the disease and deaths, the decline in vaccine-induced immunity over time and the continuing emergence of new viral variants or mutations underscore the need for an alternative strategy for developing broad-spectrum host-mediated therapeutics against SARS-CoV-2. A key feature of severe COVID-19 is dysregulated innate immune signaling, culminating in a high expression of numerous pro-inflammatory cytokines and chemokines and a lack of antiviral interferons (IFNs), particularly type I (alpha and beta) and type III (lambda). As a natural host defense, the myeloid differentiation primary response protein, MyD88, plays pivotal roles in innate and acquired immune responses via the signal transduction pathways of Toll-like receptors (TLRs), a type of pathogen recognition receptors (PRRs). However, recent studies have highlighted that infection with viruses upregulates MyD88 expression and impairs the host antiviral response by negatively regulating type I IFN. Galectin-3 (Gal3), another key player in viral infections, has been shown to modulate the host immune response by regulating viral entry and activating TLRs, the NLRP3 inflammasome, and NF-κB, resulting in the release of pro-inflammatory cytokines and contributing to the overall inflammatory response, the so-called \"cytokine storm\". These studies suggest that the specific inhibition of MyD88 and Gal3 could be a promising therapy for COVID-19. This review presents future directions for MyD88- and Gal3-targeted antiviral drug discovery, highlighting the potential to restore host immunity in SARS-CoV-2 infections.
    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
    溃疡性结肠炎,炎症性肠病,以氧化应激和炎症状态为特征。芦丁是一种具有多种药理活性的天然黄酮类化合物,其通过高迁移率族B1(HMGB1)/Toll样受体-4(TLR4)/髓样分化原发反应蛋白88(MYD88)/核因子-kB(NF-kB)信号通路在醋酸诱导的溃疡性结肠炎中的作用有待探索。
    将四个实验组分为对照组,芦丁组:100mg/kg/天口服芦丁治疗10天,乙酸(AA)组:给予结肠内滴注AA诱导溃疡性结肠炎,乙酸与芦丁处理(AA/芦丁)组。
    乙酸引起结肠重量/长度比的显着增加,并引起结肠组织病理学变化,导致结肠组织病理学评分显著上升。乙酸显示LDH和CRP血清水平以及TOS结肠水平显着升高,与对照组相比,TAS结肠含量显着下降。此外,AA诱导HMGB1/TLR4/MYD88/NF-kB信号通路的激活.芦丁显示结肠重量/长度比显著降低,改善了AA引起的结肠组织病理学变化,结肠组织病理学评分明显下降。芦丁显示血清LDH显著降低,与AA组相比,CRP水平以及结肠TOS含量。芦丁抑制HMGB1/TLR4/MYD88/NF-kB信号通路的结肠活化。
    芦丁通过靶向HMGB1/TLR4/MYD88/NF-kB信号通路,可能成为抗AA诱导的溃疡性结肠炎的有前途的保生剂。
    UNASSIGNED: Ulcerative colitis, an inflammatory bowel disease, is characterized by a status of oxidative stress and inflammation. Rutin is a natural flavonoid with many pharmacological activities and its role in acetic acid-induced ulcerative colitis through the high mobility group B1 (HMGB1)/ toll-like receptor-4 (TLR4)/ myeloid differentiation primary response protein 88 (MYD88)/ nuclear factor-kB (NF-kB) signaling pathway needs to be explored.
    UNASSIGNED: Four experimental groups were divided into control group, rutin group: treated with 100 mg/kg/day rutin orally for 10 days, acetic acid (AA) group: given intracolonic instillation of AA to induce ulcerative colitis, and acetic acid with rutin treatment (AA/Rutin) group.
    UNASSIGNED: Acetic acid caused a marked increase in the colon weight/length ratio and induced colonic histopathological changes, leading to a marked rise in the colonic histopathological scores. Acetic acid exhibited a significant rise in LDH and CRP serum levels as well as TOS colonic levels, accompanied by a marked decline in TAS colonic contents compared to the control group. Moreover, AA-induced activation of the HMGB1/TLR4/MYD88/NF-kB signaling pathway. Rutin demonstrated a significant decrease in the colon weight/length ratio, ameliorated the colonic histopathological changes induced by AA, and exhibited a marked decline in the colonic histopathological scores. Rutin showed a significant decrease in serum LDH, and CRP levels as well as colonic TOS contents when compared with the AA group. Rutin suppressed the colonic activation of the HMGB1/TLR4/MYD88/NF-kB signaling pathway.
    UNASSIGNED: Rutin could be a promising coloprotective agent against AA-induced ulcerative colitis by targeting the HMGB1/TLR4/MYD88/NF-kB signaling pathway.
    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
    炎症是身体对损伤的反应,这取决于许多监管因素。其中,miRNAs因其在多个水平上调节炎症基因表达的作用而备受关注。特别是,miR-21在炎症反应期间上调,据报道通过下调促炎介质参与炎症的消退。包括MyD88。在这里,我们在6-merHA寡糖诱导人软骨细胞炎症的体外模型中评估了miR-21对TLR-4/MyD88通路的调节作用.软骨细胞暴露于6-merHA诱导了TLR4/MyD88通路的激活,最终导致NF-kB激活。miR-21,TLR-4,MyD88,NLRP3炎性体的变化,IL-29,Caspase1,MMP-9,iNOS,实时定量PCR检测6聚体HA刺激的软骨细胞的COX-2mRNA表达。TLR-4,MyD88,NLRP3炎性体的蛋白质量,p-ERK1/2,p-AKT,IL-29,caspase1,MMP-9,p-NK-kBp65亚基,和IKB-a已通过ELISA试剂盒进行了评估。NO和PGE2水平已通过比色法和ELISA试剂盒测定,分别。HA寡糖诱导上述参数的表达显著增加,包括NF-kB活性。miR-21模拟物的使用减弱了MyD88表达水平和下游效应物。相反,用miR-21抑制剂治疗诱导相反效果。有趣的是,使用MyD88siRNA证实MyD88作为miR-21的作用靶点。我们的结果表明,miR-21表达可能会增加,以减少炎症反应。瞄准MyD88.
    Inflammation is the body\'s response to injuries, which depends on numerous regulatory factors. Among them, miRNAs have gained much attention for their role in regulating inflammatory gene expression at multiple levels. In particular, miR-21 is up-regulated during the inflammatory response and reported to be involved in the resolution of inflammation by down-regulating pro-inflammatory mediators, including MyD88. Herein, we evaluated the regulatory effects of miR-21 on the TLR-4/MyD88 pathway in an in vitro model of 6-mer HA oligosaccharides-induced inflammation in human chondrocytes. The exposition of chondrocytes to 6-mer HA induced the activation of the TLR4/MyD88 pathway, which culminates in NF-kB activation. Changes in miR-21, TLR-4, MyD88, NLRP3 inflammasome, IL-29, Caspase1, MMP-9, iNOS, and COX-2 mRNA expression of 6-mer HA-stimulated chondrocytes were examined by qRT-PCR. Protein amounts of TLR-4, MyD88, NLRP3 inflammasome, p-ERK1/2, p-AKT, IL-29, caspase1, MMP-9, p-NK-kB p65 subunit, and IKB-a have been evaluated by ELISA kits. NO and PGE2 levels have been assayed by colorimetric and ELISA kits, respectively. HA oligosaccharides induced a significant increase in the expression of the above parameters, including NF-kB activity. The use of a miR-21 mimic attenuated MyD88 expression levels and the downstream effectors. On the contrary, treatment with a miR-21 inhibitor induced opposite effects. Interestingly, the use of a MyD88 siRNA confirmed MyD88 as the target of miR-21 action. Our results suggest that miR-21 expression could increase in an attempt to reduce the inflammatory response, targeting MyD88.
    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
    胰腺导管腺癌(PDAC)是一种高致命性疾病,生存率低,主要是由于基于吉西他滨(Gem)的化疗效果有限,以及获得化疗抗性。Aronia浆果提取物(ABEs),富含酚类成分,最近已经认识到它们的抗癌特性以及它们在帮助克服各种癌症中的化学抗性方面令人鼓舞的潜力。在本研究中,我们探索了ABE在PDAC中克服宝石抗性的潜力,并确定了负责其抗癌活性的特定生长调节途径。通过对吉西他滨耐药(Gem-R)细胞的一系列体外实验,我们阐明了Gem和ABE治疗之间的协同相互作用。使用高级转录组学分析和网络药理学,我们揭示了在Gem-RPDAC细胞中与ABE的化学耐药和潜在治疗靶点相关的关键分子途径.随后,细胞培养研究的结果在患者来源的3D肿瘤类器官(PDO)中得到验证.ABE和Gem的联合治疗对细胞活力显示出显著的协同作用和抗癌作用,扩散,迁移,和侵入Gem-R细胞。转录组分析显示NF-κb信号通路与Gem-R之间存在相关性(p<0.05),表现出MYD88的明显上调。此外,MYD88与TCGA队列中PDAC患者的总生存率显著相关(HR=1.58,p<0.05)。MYD88/NF-κb途径通过潜在上调外排转运蛋白如P-糖蛋白(P-gp)来促进化学抗性。我们的发现表明,与ABE联合治疗通过靶向MYD88和减少P-gp表达来抑制NF-κb途径以克服宝石抗性。最后,在PDO中,联合治疗被证明在减少其数量和大小方面非常有效(p<0.05).我们的研究为ABE通过靶向MYD88/NF-κb/P-gp轴克服PDAC细胞中宝石耐药性的能力提供了以前未被认可的见解。因此提供了一种安全且具有成本效益的辅助治疗策略,以改善PDAC的治疗结果.
    Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with poor survival rates, primarily due to the limited effectiveness of gemcitabine (Gem)-based chemotherapy, as well as the acquisition of chemotherapeutic resistance. Aronia berry extracts (ABEs), abundant in phenolic constituents, have been recently recognized for their anticancer properties as well as their encouraging potential to help overcome chemoresistance in various cancers. In the present study, we explored ABE\'s potential to overcome Gem resistance in PDAC and identify specific growth regulatory pathways responsible for its anticancer activity. Through a series of in vitro experiments in gemcitabine-resistant (Gem-R) cells, we elucidated the synergistic interactions between Gem and ABE treatments. Using advanced transcriptomic analysis and network pharmacology, we revealed key molecular pathways linked to chemoresistance and potential therapeutic targets of ABE in Gem-R PDAC cells. Subsequently, the findings from cell culture studies were validated in patient-derived 3D tumor organoids (PDOs). The combination treatment of ABE and Gem demonstrated significant synergism and anticancer effects on cell viability, proliferation, migration, and invasion in Gem-R cells. Transcriptomic analysis revealed a correlation between the NF-Κb signaling pathway and Gem-R (p < 0.05), exhibiting a marked upregulation of MYD88. Additionally, MYD88 exhibited a significant correlation with the overall survival rates in patients with PDAC patients in the TCGA cohort (HR = 1.58, p < 0.05). The MYD88/NF-Κb pathway contributes to chemoresistance by potentially upregulating efflux transporters like P-glycoprotein (P-gp). Our findings revealed that the combined treatment with ABE suppressed the NF-Κb pathway by targeting MYD88 and reducing P-gp expression to overcome Gem resistance. Lastly, the combination therapy proved highly effective in PDOs in reducing both their number and size (p < 0.05). Our study offers previously unrecognized insights into the ability of ABE to overcome Gem resistance in PDAC cells through its targeting of the MYD88/NF-κb/P-gp axis, hence providing a safe and cost-effective adjunctive therapeutic strategy to improve treatment outcomes in PDAC.
    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
    查菲埃里希菌在单核细胞或巨噬细胞内感染和增殖,并导致人单核细胞埃里希菌病(HME),一种正在出现的威胁生命的蜱传人畜共患病。内化后,E.Chaffeensis驻留在专门的膜结合内含物中,E.Chaffeensis-containingvesicles(ECVs),逃避宿主细胞先天免疫反应并获得营养。然而,宿主细胞在ECV中抑制Chaffeensis生长的机制仍然未知。在这里,我们证明宿主细胞识别E.chaffeensisEch_1067,一种青霉素结合蛋白,然后上调PIT1的表达,PIT1是一种磷酸盐转运蛋白,将磷酸盐从ECV转运到细胞质中以抑制细菌生长。我们发现,宿主细胞上调PIT1的表达时,使用转录组测序,qRT-PCR和蛋白质印迹,使用共聚焦显微镜将PIT1定位于感染的THP-1细胞的ECV膜上。使用shRNA的PIT1的沉默增强了查夫森大肠杆菌的细胞内生长。最后,我们发现,使用重组蛋白刺激和siRNA沉默,通过MyD88-NF-κB途径诱导PIT1表达上调。我们的发现加深了对宿主细胞抑制细菌细胞内生长的先天免疫反应的理解,并促进了HME新疗法的开发。
    Ehrlichia chaffeensis infects and proliferates inside monocytes or macrophages and causes human monocytic ehrlichiosis (HME), an emerging life-threatening tick-borne zoonosis. After internalization, E. chaffeensis resides in specialized membrane-bound inclusions, E. chaffeensis-containing vesicles (ECVs), to evade from host cell innate immune responses and obtain nutrients. However, mechanisms exploited by host cells to inhibit E. chaffeensis growth in ECVs are still largely unknown. Here we demonstrate that host cells recognize E. chaffeensis Ech_1067, a penicillin-binding protein, and then upregulate the expression of PIT1, which is a phosphate transporter and transports phosphate from ECVs to the cytosol to inhibit bacterial growth. We found that host cells upregulate the PIT1 expression upon E. chaffeensis infection using transcriptome sequencing, qRT-PCR and Western blotting, and PIT1 is localized on the ECV membrane in infected THP-1 cells using confocal microscopy. Silence of PIT1 using shRNA enhances E. chaffeensis intracellular growth. Finally, we found that E. chaffeensis Ech_1067 induces the upregulation of PIT1 expression through the MyD88-NF-κB pathway using recombinant protein for stimulation and siRNA for silence. Our findings deepen the understanding of the innate immune responses of host cells to inhibit bacterial intracellular growth and facilitate the development of new therapeutics for HME.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号