Cellular Reprogramming

细胞重编程
  • 文章类型: Journal Article
    在复杂的肿瘤微环境中,肿瘤相关巨噬细胞(TAMs)作为一种普遍存在的细胞成分,对致癌过程产生深远的影响。肝细胞癌(HCC)的微环境的特点是TAM的明显浸润,强调了它们在调节疾病轨迹中的关键作用。在不断发展的肝癌治疗范式中,代谢途径的战略重新编程为干预提供了一个有希望的途径,在科学界引起了越来越多的兴趣。以前的研究主要集中在阐明癌细胞中代谢重编程的机制,而没有给予足够的关注来理解TAM代谢重编程。特别是脂质代谢,影响HCC的进展。在这篇评论文章中,我们打算阐明TAM如何通过多种途径发挥其调节作用,如E2F1-E2F2-CPT2,LKB1-AMPK,和mTORC1-SREBP,并通过巩固TAM脂质摄取的各种研究,讨论TAM与这些过程的相关性以及HCC进展中相关途径的特征,storage,合成,和分解代谢。我们希望我们的总结可以描述TAM脂质代谢重编程对HCC进展的具体机制的影响,并为HCC的未来研究和新治疗策略的开发提供有用的信息。
    In the intricate landscape of the tumor microenvironment, tumor-associated macrophages (TAMs) emerge as a ubiquitous cellular component that profoundly affects the oncogenic process. The microenvironment of hepatocellular carcinoma (HCC) is characterized by a pronounced infiltration of TAMs, underscoring their pivotal role in modulating the trajectory of the disease. Amidst the evolving therapeutic paradigms for HCC, the strategic reprogramming of metabolic pathways presents a promising avenue for intervention, garnering escalating interest within the scientific community. Previous investigations have predominantly focused on elucidating the mechanisms of metabolic reprogramming in cancer cells without paying sufficient attention to understanding how TAM metabolic reprogramming, particularly lipid metabolism, affects the progression of HCC. In this review article, we intend to elucidate how TAMs exert their regulatory effects via diverse pathways such as E2F1-E2F2-CPT2, LKB1-AMPK, and mTORC1-SREBP, and discuss correlations of TAMs with these processes and the characteristics of relevant pathways in HCC progression by consolidating various studies on TAM lipid uptake, storage, synthesis, and catabolism. It is our hope that our summary could delineate the impact of specific mechanisms underlying TAM lipid metabolic reprogramming on HCC progression and provide useful information for future research on HCC and the development of new treatment strategies.
    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
    原理:代谢的协同重编程主导神经母细胞瘤(NB)的进展。基于阐明代谢重编程的分子机制,开发具有分层指导的NB治疗选择的个性化风险预测方法具有重要的临床意义。方法:采用基于机器学习的多步骤程序,在单细胞和代谢物通量维度阐明了代谢重编程驱动NB恶性进展的协同机制.随后,我们开发了一种有前景的代谢重编程相关预后特征(MPS)和基于MPS分层的个体化治疗方法,并使用临床前模型进行了进一步独立验证.结果:MPS鉴定的MPS-INB显示出明显高于MPS-II对应物的代谢重编程活性。与目前的临床特征相比,MPS显示出更高的准确性[AUC:0.915vs.0.657(MYCN),0.713(INSS阶段),和0.808(INRG分层)]预测预后。AZD7762和依托泊苷被确定为针对MPS-I和IINB的有效治疗剂,分别。随后的生物学测试表明AZD7762基本上抑制了生长,迁移,MPS-INB细胞的侵袭,比MPS-II细胞更有效。相反,依托泊苷对MPS-Ⅱ型NB细胞有较好的治疗作用。更令人鼓舞的是,AZD7762和依托泊苷显著抑制体内皮下肿瘤发生,扩散,MPS-I和MPS-II样本中的肺转移,分别;从而延长荷瘤小鼠的生存期。机械上,AZD7762和依托泊苷诱导的MPS-I和MPS-II细胞凋亡,分别,通过线粒体依赖性途径;MPS-INB通过谷氨酸代谢成瘾和乙酰辅酶A抵抗依托泊苷诱导的细胞凋亡。MPS-INB进展受多种代谢重编程驱动因素的推动,包括多药耐药,免疫抑制和促进肿瘤的炎症微环境。免疫学,MPS-INB通过MIF和THBS信号通路抑制免疫细胞。代谢,MPS-INB细胞的恶性增殖得到了重新编程的谷氨酸代谢的显著支持,三羧酸循环,尿素循环,等。此外,MPS-INB细胞表现出独特的肿瘤促进发育谱系和自我沟通模式,通过发育和自我通讯激活的致癌信号通路增强证明了这一点。结论:本研究为代谢重编程介导的NB恶性进展的分子机制提供了深刻的见解。它还揭示了在新的精确风险预测方法的指导下开发靶向药物,这可能有助于明显改善NB的治疗策略。
    Rationale: Synergic reprogramming of metabolic dominates neuroblastoma (NB) progression. It is of great clinical implications to develop an individualized risk prognostication approach with stratification-guided therapeutic options for NB based on elucidating molecular mechanisms of metabolic reprogramming. Methods: With a machine learning-based multi-step program, the synergic mechanisms of metabolic reprogramming-driven malignant progression of NB were elucidated at single-cell and metabolite flux dimensions. Subsequently, a promising metabolic reprogramming-associated prognostic signature (MPS) and individualized therapeutic approaches based on MPS-stratification were developed and further validated independently using pre-clinical models. Results: MPS-identified MPS-I NB showed significantly higher activity of metabolic reprogramming than MPS-II counterparts. MPS demonstrated improved accuracy compared to current clinical characteristics [AUC: 0.915 vs. 0.657 (MYCN), 0.713 (INSS-stage), and 0.808 (INRG-stratification)] in predicting prognosis. AZD7762 and etoposide were identified as potent therapeutics against MPS-I and II NB, respectively. Subsequent biological tests revealed AZD7762 substantially inhibited growth, migration, and invasion of MPS-I NB cells, more effectively than that of MPS-II cells. Conversely, etoposide had better therapeutic effects on MPS-II NB cells. More encouragingly, AZD7762 and etoposide significantly inhibited in-vivo subcutaneous tumorigenesis, proliferation, and pulmonary metastasis in MPS-I and MPS-II samples, respectively; thereby prolonging survival of tumor-bearing mice. Mechanistically, AZD7762 and etoposide-induced apoptosis of the MPS-I and MPS-II cells, respectively, through mitochondria-dependent pathways; and MPS-I NB resisted etoposide-induced apoptosis by addiction of glutamate metabolism and acetyl coenzyme A. MPS-I NB progression was fueled by multiple metabolic reprogramming-driven factors including multidrug resistance, immunosuppressive and tumor-promoting inflammatory microenvironments. Immunologically, MPS-I NB suppressed immune cells via MIF and THBS signaling pathways. Metabolically, the malignant proliferation of MPS-I NB cells was remarkably supported by reprogrammed glutamate metabolism, tricarboxylic acid cycle, urea cycle, etc. Furthermore, MPS-I NB cells manifested a distinct tumor-promoting developmental lineage and self-communication patterns, as evidenced by enhanced oncogenic signaling pathways activated with development and self-communications. Conclusions: This study provides deep insights into the molecular mechanisms underlying metabolic reprogramming-mediated malignant progression of NB. It also sheds light on developing targeted medications guided by the novel precise risk prognostication approaches, which could contribute to a significantly improved therapeutic strategy for NB.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 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
    视网膜神经节细胞(RGC)将视网膜连接到大脑。RGC的轴突和树突的适当发育是这些细胞充当投射神经元以向大脑传递视觉信息的基础。这项研究的目的是研究Shtn1(编码shootin1)在RGC神经突发育中的功能。
    免疫荧光(IF)用于表征标记基因的表达模式。体外直接体细胞重编程系统用于产生RGC样神经元(iRGC),随后用于研究Shtn1的功能。短发夹RNA(shRNA)用于敲除Shtn1,并且Shtn1的编码序列(CDS)用于过表达该基因。慢病毒用于将shRNA或CDS递送到iRGC中。膜片钳技术用于测量iRGC的电生理特性。RNA测序(RNA-seq)用于检查转录组表达。
    使用IF,我们证明,在RGC活跃发育并调节其神经突与上游和下游神经元的连接期间,shootin1在RGC中明显表达。使用iRGC系统,我们证明了Shtn1促进神经突的生长和复杂性,从而促进电生理成熟,iRGC的。RNA-seq分析表明,Shtn1也可能调控iRGC中的基因表达和神经发生。
    Shtn1促进RGC神经突发育。这些发现提高了我们对控制RGC神经突发育的分子机制的理解,并可能有助于优化未来的RGC再生方法。
    UNASSIGNED: Retinal ganglion cells (RGCs) connect the retina to the brain. Proper development of the axons and dendrites of RGCs is the basis for these cells to function as projection neurons to deliver visual information to the brain. The purpose of this study was to investigate the function of Shtn1 (which encodes shootin1) in RGC neurite development.
    UNASSIGNED: Immunofluorescence (IF) was used to characterize the expression pattern of marker genes. An in vitro direct somatic cell reprogramming system was used to generate RGC-like neurons (iRGCs), which was subsequently used to study the function of Shtn1. Short-hairpin RNAs (shRNAs) were used to knock down Shtn1, and the coding sequence (CDS) of Shtn1 was used to overexpress the gene. Lentiviruses were used to deliver shRNAs or CDSs into iRGCs. The patch clamp technique was used to measure the electrophysiological properties of the iRGCs. RNA sequencing (RNA-seq) was used to examine transcriptome expression.
    UNASSIGNED: Using IF, we demonstrated that shootin1 is distinctively expressed in RGCs during the period in which RGCs actively develop and adjust the connections of their neurites with upstream and downstream neurons. Using the iRGC system, we demonstrated that Shtn1 promotes the growth and complexity of neurites and thus the electrophysiological maturation, of iRGCs. RNA-seq analyses showed that Shtn1 may also regulate gene expression and neurogenesis in iRGCs.
    UNASSIGNED: Shtn1 promotes RGC neurite development. These findings improve our understanding of the molecular machinery governing RGC neurite development and may help to optimize future RGC regeneration methods.
    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
    背景:成年哺乳动物心脏的再生能力有限。许多研究已经探索了成年心肌细胞细胞周期退出的机制。这项翻译研究评估了rhCHK1(重组人检查点激酶1)对猪缺血/再灌注损伤后心肌细胞存活和增殖以及心肌修复的影响及其潜在机制。
    结果:心肌内注射包裹在水凝胶中的rhCHK1蛋白(1mg/kg)在缺血/再灌注损伤后3天刺激心肌细胞增殖并降低心脏炎症反应,改善心功能,减轻心室重构,并在缺血/再灌注损伤后28天减少梗死面积。机械上,多组学测序分析表明rhCHK1治疗后糖酵解和mTOR(哺乳动物雷帕霉素靶蛋白)途径的富集.免疫共沉淀(Co-IP)实验和蛋白质对接预测显示CHK1(检查点激酶1)直接结合并激活PKM2(丙酮酸激酶同工型M2)的丝氨酸37(S37)和酪氨酸105(Y105)位点以促进代谢重编程。我们进一步构建了敲除不同CHK1和PKM2氨基酸结构域的质粒,并将其转染到人胚胎肾293T(HEK293T)细胞中进行CO-IP实验。结果显示CHK1的1-265结构域直接结合PKM2的157-400个氨基酸。此外,hiPSC-CM(人iPS细胞衍生的心肌细胞)的体外和体内实验均表明,CHK1通过激活PKM2C结构域介导的心脏代谢重编程刺激心肌细胞更新和心脏修复。
    结论:本研究表明,CHK1的1-265个氨基酸结构域与PKM2的157-400结构域结合,激活PKM2介导的代谢重编程,促进成年猪缺血再灌注损伤后心肌细胞增殖和心肌修复。
    BACKGROUND: The regenerative capacity of the adult mammalian hearts is limited. Numerous studies have explored mechanisms of adult cardiomyocyte cell-cycle withdrawal. This translational study evaluated the effects and underlying mechanism of rhCHK1 (recombinant human checkpoint kinase 1) on the survival and proliferation of cardiomyocyte and myocardial repair after ischemia/reperfusion injury in swine.
    RESULTS: Intramyocardial injection of rhCHK1 protein (1 mg/kg) encapsulated in hydrogel stimulated cardiomyocyte proliferation and reduced cardiac inflammation response at 3 days after ischemia/reperfusion injury, improved cardiac function and attenuated ventricular remodeling, and reduced the infarct area at 28 days after ischemia/reperfusion injury. Mechanistically, multiomics sequencing analysis demonstrated enrichment of glycolysis and mTOR (mammalian target of rapamycin) pathways after rhCHK1 treatment. Co-Immunoprecipitation (Co-IP) experiments and protein docking prediction showed that CHK1 (checkpoint kinase 1) directly bound to and activated the Serine 37 (S37) and Tyrosine 105 (Y105) sites of PKM2 (pyruvate kinase isoform M2) to promote metabolic reprogramming. We further constructed plasmids that knocked out different CHK1 and PKM2 amino acid domains and transfected them into Human Embryonic Kidney 293T (HEK293T) cells for CO-IP experiments. Results showed that the 1-265 domain of CHK1 directly binds to the 157-400 amino acids of PKM2. Furthermore, hiPSC-CM (human iPS cell-derived cardiomyocyte) in vitro and in vivo experiments both demonstrated that CHK1 stimulated cardiomyocytes renewal and cardiac repair by activating PKM2 C-domain-mediated cardiac metabolic reprogramming.
    CONCLUSIONS: This study demonstrates that the 1-265 amino acid domain of CHK1 binds to the 157-400 domain of PKM2 and activates PKM2-mediated metabolic reprogramming to promote cardiomyocyte proliferation and myocardial repair after ischemia/reperfusion injury in adult pigs.
    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
    前列腺癌(PCa)仍然是美国男性死亡的主要原因,由于对控制疾病发生的潜在生物过程的理解有限,转移性和复发性疾病构成了重大的治疗挑战,休眠,和进步。事实证明,PCa细胞系的常规使用不足以阐明驱动PCa癌变的复杂分子机制。阻碍有效治疗的发展。为了解决这个差距,已经开发了患者来源的原代细胞培养物,并且在揭示每个个体中PCa特有的病理生理复杂性方面发挥着关键作用,为转化研究提供有价值的见解。本文探讨了条件重编程(CR)细胞培养方法的应用,展示了其快速有效培养患者来源的正常细胞和肿瘤细胞的能力。CR策略有助于原代细胞获得干细胞特性,精确地概括了PCa的人类病理生理学。这种细致入微的理解使得能够识别新的疗法。具体来说,我们的讨论包括CR细胞在阐明PCa起始和进展中的效用,揭示转移性PCa的分子发病机制,解决健康差距,推进个性化医疗。再加上肿瘤类器官方法和患者来源的异种移植物(PDX),CR细胞为理解癌症生物学提供了一个有希望的途径,探索新的治疗方式,在PCa的背景下推进精准医学。这些方法已用于两个NCI计划(PDMR:患者衍生的模型库;HCMI:人类癌症模型计划)。
    Prostate cancer (PCa) remains a leading cause of mortality among American men, with metastatic and recurrent disease posing significant therapeutic challenges due to a limited comprehension of the underlying biological processes governing disease initiation, dormancy, and progression. The conventional use of PCa cell lines has proven inadequate in elucidating the intricate molecular mechanisms driving PCa carcinogenesis, hindering the development of effective treatments. To address this gap, patient-derived primary cell cultures have been developed and play a pivotal role in unraveling the pathophysiological intricacies unique to PCa in each individual, offering valuable insights for translational research. This review explores the applications of the conditional reprogramming (CR) cell culture approach, showcasing its capability to rapidly and effectively cultivate patient-derived normal and tumor cells. The CR strategy facilitates the acquisition of stem cell properties by primary cells, precisely recapitulating the human pathophysiology of PCa. This nuanced understanding enables the identification of novel therapeutics. Specifically, our discussion encompasses the utility of CR cells in elucidating PCa initiation and progression, unraveling the molecular pathogenesis of metastatic PCa, addressing health disparities, and advancing personalized medicine. Coupled with the tumor organoid approach and patient-derived xenografts (PDXs), CR cells present a promising avenue for comprehending cancer biology, exploring new treatment modalities, and advancing precision medicine in the context of PCa. These approaches have been used for two NCI initiatives (PDMR: patient-derived model repositories; HCMI: human cancer models initiatives).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Interview
    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
    狼疮性肾炎(LN)是系统性红斑狼疮(SLE)的严重和常见表现,经常被确定为预后不良。巨噬细胞在其发病机制中起重要作用。不同的巨噬细胞亚型对狼疮受累的肾脏有不同的影响。基于它们的起源,巨噬细胞可分为单核细胞衍生的巨噬细胞(MoMacs)和组织驻留的巨噬细胞(TrMacs)。在肾炎期间,TrMacs发展出一种混合的促炎和抗炎功能表型,因为它们在受细胞因子刺激时不分泌精氨酸酶或一氧化氮(NO)。这些混合表型巨噬细胞的浸润与免疫复合物和暴露于循环炎症介质引起的持续损伤有关,这是无法解决炎症的迹象。另一方面,MoMac在细胞因子刺激下分化成M1或M2细胞。M1巨噬细胞是促炎的,分泌促炎细胞因子,而M2主要表型本质上是抗炎和促进组织修复。相反,响应于细胞因子刺激,MoMac经历分化成M1或M2细胞。M1巨噬细胞被认为是促炎细胞和分泌促炎介质,而M2主要表型主要是抗炎和促进组织修复。此外,基于细胞因子的表达,M2巨噬细胞可进一步分为M2a、M2b,和M2c表型。M2a和M2c具有抗炎作用,参与组织修复,而M2b细胞具有免疫调节和促炎特性。Further,记忆巨噬细胞也在LN的发展中起作用。研究表明,巨噬细胞的极化受多种代谢途径控制,比如糖酵解,磷酸戊糖途径,脂肪酸氧化,鞘脂代谢,三羧酸循环,和精氨酸代谢。这些代谢途径的变化可以受到鱼油等物质的调节,多烯磷脂酰胆碱,牛磺酸,富马酸,二甲双胍,和沙丁胺醇,抑制巨噬细胞的M1极化,促进M2极化,从而减轻LN。
    Lupus nephritis (LN) is a severe and common manifestation of systemic lupus erythematosus (SLE) that is frequently identified with a poor prognosis. Macrophages play an important role in its pathogenesis. Different macrophage subtypes have different effects on lupus-affected kidneys. Based on their origin, macrophages can be divided into monocyte-derived macrophages (MoMacs) and tissue-resident macrophages (TrMacs). During nephritis, TrMacs develop a hybrid pro-inflammatory and anti-inflammatory functional phenotype, as they do not secrete arginase or nitric oxide (NO) when stimulated by cytokines. The infiltration of these mixed-phenotype macrophages is related to the continuous damage caused by immune complexes and exposure to circulating inflammatory mediators, which is an indication of the failure to resolve inflammation. On the other hand, MoMacs differentiate into M1 or M2 cells under cytokine stimulation. M1 macrophages are pro-inflammatory and secrete pro-inflammatory cytokines, while the M2 main phenotype is essentially anti-inflammatory and promotes tissue repair. Conversely, MoMacs undergo differentiation into M1 or M2 cells in response to cytokine stimulation. M1 macrophages are considered pro-inflammatory cells and secrete pro-inflammatory mediators, whereas the M2 main phenotype is primarily anti-inflammatory and promotes tissue repair. Moreover, based on cytokine expression, M2 macrophages can be further divided into M2a, M2b, and M2c phenotypes. M2a and M2c have anti-inflammatory effects and participate in tissue repair, while M2b cells have immunoregulatory and pro-inflammatory properties. Further, memory macrophages also have a role in the advancement of LN. Studies have demonstrated that the polarization of macrophages is controlled by multiple metabolic pathways, such as glycolysis, the pentose phosphate pathway, fatty acid oxidation, sphingolipid metabolism, the tricarboxylic acid cycle, and arginine metabolism. The changes in these metabolic pathways can be regulated by substances such as fish oil, polyenylphosphatidylcholine, taurine, fumaric acid, metformin, and salbutamol, which inhibit M1 polarization of macrophages and promote M2 polarization, thereby alleviating LN.
    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
    胰腺癌是致命的恶性肿瘤之一,死亡率很高,目前治疗方法很少。胰腺癌的肿瘤微环境(TME),以纤维化和癌症相关成纤维细胞(CAF)的存在为特征,对肿瘤的进展和对治疗的抵抗力都有重要影响。工程细胞外囊泡(EV)领域的最新进展为胰腺癌的靶向治疗提供了新途径。本研究旨在开发用于CAF的靶向重编程和调节胰腺癌中的TME的工程化EV。将从骨髓间充质干细胞(BMSCs)获得的EV加载miR-138-5p和抗纤维化剂吡非尼酮(PFD),并用整合素α5靶向肽(命名为IEVs-PFD/138)进行表面修饰,以重新编程CAFs并抑制其促肿瘤作用。整合素α5靶向肽修饰增强了EV的CAF靶向能力。miR-138-5p直接抑制FERMT2-TGFBR1复合物的形成,抑制TGF-β信号通路的激活。此外,miR-138-5p通过直接靶向FERMT2-PYCR1复合物抑制脯氨酸介导的胶原合成。EV中miR-138-5p和PFD的组合协同促进CAF重编程并抑制CAF的促癌作用。使用富含原位基质和患者来源的异种移植小鼠模型的临床前实验产生了有希望的结果。特别是,IEVs-PFD/138有效地重新编程CAF和改造TME,导致肿瘤压力下降,增强吉西他滨灌注,肿瘤缺氧改善,癌细胞对化疗的敏感性更高。因此,本研究制定的策略可以改善化疗结局.利用IEV-PFD/138作为靶向治疗剂来调节CAF和TME代表了胰腺癌的有希望的治疗方法。
    Pancreatic cancer is one of the deadly malignancies with a significant mortality rate and there are currently few therapeutic options for it. The tumor microenvironment (TME) in pancreatic cancer, distinguished by fibrosis and the existence of cancer-associated fibroblasts (CAFs), exerts a pivotal influence on both tumor advancement and resistance to therapy. Recent advancements in the field of engineered extracellular vesicles (EVs) offer novel avenues for targeted therapy in pancreatic cancer. This study aimed to develop engineered EVs for the targeted reprogramming of CAFs and modulating the TME in pancreatic cancer. EVs obtained from bone marrow mesenchymal stem cells (BMSCs) were loaded with miR-138-5p and the anti-fibrotic agent pirfenidone (PFD) and subjected to surface modification with integrin α5-targeting peptides (named IEVs-PFD/138) to reprogram CAFs and suppress their pro-tumorigenic effects. Integrin α5-targeting peptide modification enhanced the CAF-targeting ability of EVs. miR-138-5p directly inhibited the formation of the FERMT2-TGFBR1 complex, inhibiting TGF-β signaling pathway activation. In addition, miR-138-5p inhibited proline-mediated collagen synthesis by directly targeting the FERMT2-PYCR1 complex. The combination of miR-138-5p and PFD in EVs synergistically promoted CAF reprogramming and suppressed the pro-cancer effects of CAFs. Preclinical experiments using the orthotopic stroma-rich and patient-derived xenograft mouse models yielded promising results. In particular, IEVs-PFD/138 effectively reprogrammed CAFs and remodeled TME, which resulted in decreased tumor pressure, enhanced gemcitabine perfusion, tumor hypoxia amelioration, and greater sensitivity of cancer cells to chemotherapy. Thus, the strategy developed in this study can improve chemotherapy outcomes. Utilizing IEVs-PFD/138 as a targeted therapeutic agent to modulate CAFs and the TME represents a promising therapeutic approach for pancreatic cancer.
    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
    上呼吸道鳞状细胞癌(UASCC)是一种常见的侵袭性恶性肿瘤,几乎没有有效的治疗选择。这里,我们研究了这种癌症的氨基酸代谢,令人惊讶的是,UASCC在所有人类癌症中表现出最高的蛋氨酸水平,由其运输车LAT1驱动。我们表明LAT1在UASCC中也以最高水平表达,由UASCC特异性启动子和增强子转录激活,它们由SCC主调节剂TP63/KLF5/SREBF1直接共同调节。出乎意料的是,无偏生物信息学筛选将EZH2鉴定为LAT1-蛋氨酸途径下游的最重要靶标,直接将蛋氨酸代谢与表观基因组重编程联系起来。重要的是,这种级联反应对于UASCC患者来源的肿瘤类器官的存活和增殖是必不可少的。此外,LAT1表达与细胞对LAT1-甲硫氨酸-EZH2轴抑制的敏感性密切相关。值得注意的是,这种独特的LAT1-蛋氨酸-EZH2级联反应可以通过体内药理学方法或饮食干预有效靶向。总之,这项工作映射了表观基因组重编程与蛋氨酸代谢之间的独特机制串扰,确立了其在UASCC生物学中的生物学意义,并确定了一种独特的肿瘤特异性脆弱性,可以在药理和饮食上利用。
    Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号