Metabolic Reprogramming

代谢重编程
  • 文章类型: Journal Article
    越来越多的证据支持细胞竞争,多细胞生物中至关重要的选择和质量控制机制,参与肿瘤发生和发展;然而,细胞竞争与肿瘤耐药性之间的相关机制贡献仍不明确.在我们的研究中,基于对列维替尼耐药的肝细胞癌(HCC)细胞通过重编程能量代谢显示出明显的竞争生长优势。机械上,Lenvatinib耐药HCC细胞中BCL2相互作用蛋白3(BNIP3)介导的线粒体自噬的过度激活通过将能量产生从线粒体氧化磷酸化转移到糖酵解来促进糖酵解通量,通过调节AMP激活的蛋白激酶(AMPK)-烯醇化酶2(ENO2)信号,永久保持耐乐替尼肝癌细胞相对于敏感肝癌细胞的竞争优势。值得注意的是,BNIP3抑制显著敏化乐伐替尼在HCC中的抗肿瘤功效。我们的发现强调了BNIP3-AMPK-ENO2信号传导在通过调节能量代谢重编程维持耐来维替尼肝癌细胞的竞争性结果中的重要作用;同时,这项工作认为BNIP3是克服HCC耐药性的有希望的靶标。
    An increasing evidence supports that cell competition, a vital selection and quality control mechanism in multicellular organisms, is involved in tumorigenesis and development; however, the mechanistic contributions to the association between cell competition and tumor drug resistance remain ill-defined. In our study, based on a contructed lenvitinib-resistant hepatocellular carcinoma (HCC) cells display obvious competitive growth dominance over sensitive cells through reprogramming energy metabolism. Mechanistically, the hyperactivation of BCL2 interacting protein3 (BNIP3) -mediated mitophagy in lenvatinib-resistant HCC cells promotes glycolytic flux via shifting energy production from mitochondrial oxidative phosphorylation to glycolysis, by regulating AMP-activated protein kinase (AMPK) -enolase 2 (ENO2) signaling, which perpetually maintaining lenvatinib-resistant HCC cells\' competitive advantage over sensitive HCC cells. Of note, BNIP3 inhibition significantly sensitized the anti-tumor efficacy of lenvatinib in HCC. Our findings emphasize a vital role for BNIP3-AMPK-ENO2 signaling in maintaining the competitive outcome of lenvitinib-resistant HCC cells via regulating energy metabolism reprogramming; meanwhile, this work recognizes BNIP3 as a promising target to overcome HCC drug resistance.
    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
    癌症耐药性与代谢适应有关。已显示癌细胞在细胞死亡期间的适应中涉及乙酰化多胺。然而,目前尚缺乏探索乙酰化多胺作为潜在抗癌药物的模拟物。我们对用多柔比星(DOX)处理的人乳腺癌MCF-7细胞进行了细胞内代谢物谱分析,一种众所周知的抗癌药物。采用新颖的内部垂直管凝胶电泳辅助程序,然后进行LC-HRMS分析,以检测乙酰化多胺,例如N1-乙酰亚精胺。我们设计了一种模拟物N1-乙酰亚精胺(MINAS),它是组蛋白脱乙酰酶10(HDAC10)的已知底物。使用分子对接和分子动力学(MD)模拟来评估MINAS对HDAC10的抑制潜力。将MINAS的抑制潜力和ADMET曲线与已知的HDAC10抑制剂TubastatinA.N1-乙酰亚精胺进行了比较,聚胺的乙酰化形式,在用DOX处理的MCF-7细胞中细胞内检测到超过DMSO处理的MCF-7细胞。我们设计并策划了MINAS(PubChemCID162679241)。分子对接和MD模拟表明,MINAS(〜8.2kcal/mol)对TubastatinA(〜8.4kcal/mol)具有强大且可比的抑制潜力。MINAS和TubastatinA在HDAC10上具有相似的结合位点,包括Ser138、Ser140、Tyr183和Cys184。此外,MINAS与TubastatinA相比具有更好的ADMET曲线,具有较高的MRTD值和较低的毒性。总之,数据显示,N1-乙酰亚精胺水平在DOX诱导的乳腺癌细胞死亡过程中升高.此外,MINAS,一种N1-乙酰亚精胺模拟化合物,当与DOX等化疗联合使用时,可以作为潜在的抗癌药物进行研究。
    Cancer drug resistance is associated with metabolic adaptation. Cancer cells have been shown to implicate acetylated polyamines in adaptations during cell death. However, exploring the mimetic of acetylated polyamines as a potential anticancer drug is lacking. We performed intracellular metabolite profiling of human breast cancer MCF-7 cells treated with doxorubicin (DOX), a well known anticancer drug. A novel and in-house vertical tube gel electrophoresis assisted procedure followed by LC-HRMS analysis was employed to detect acetylated polyamines such as N1-acetylspermidine. We designed a mimetic N1-acetylspermidine (MINAS) which is a known substrate of histone deacetylase 10 (HDAC10). Molecular docking and molecular dynamics (MDs) simulations were used to evaluate the inhibitory potential of MINAS against HDAC10. The inhibitory potential and the ADMET profile of MINAS were compared to a known HDAC10 inhibitor Tubastatin A. N1-acetylspermidine, an acetylated form of polyamine, was detected intracellularly in MCF-7 cells treated with DOX over DMSO-treated MCF-7 cells. We designed and curated MINAS (PubChem CID 162679241). Molecular docking and MD simulations suggested the strong and comparable inhibitory potential of MINAS (-8.2 kcal/mol) to Tubastatin A (-8.4 kcal/mol). MINAS and Tubastatin A share similar binding sites on HDAC10, including Ser138, Ser140, Tyr183, and Cys184. Additionally, MINAS has a better ADMET profile compared to Tubastatin A, with a high MRTD value and lower toxicity. In conclusion, the data show that N1-acetylspermidine levels rise during DOX-induced breast cancer cell death. Additionally, MINAS, an N1-acetylspermidine mimetic compound, could be investigated as a potential anticancer drug when combined with chemotherapy like DOX.
    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
    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
    代谢重编程被认为是癌症的标志,使癌细胞获得细胞生长所必需的生物分子,通常以上调的糖酵解和/或脂肪酸合成相关基因为特征。转录因子叉头框M1(FOXM1)与各种癌症有关,对他们的发展做出重大贡献,包括结直肠癌(CRC),一个重大的全球健康问题。尽管FOXM1在癌症中的作用已经确立,其在CRC的Warburg效应和脂肪酸生物合成中的具体参与尚不清楚.我们分析了癌症基因组图谱(TCGA)结肠腺癌和直肠腺癌(COADREAD)数据集,以得出FOXM1和多个基因之间的表达水平与基于FOXM1表达的生存预后的相关性。使用两种人类CRC细胞系,HT29和HCT116,我们进行了RNAi或质粒转染程序,接着是一系列的化验,包括RNA提取,定量实时聚合酶链反应,蛋白质印迹分析,细胞代谢测定,和免疫荧光分析。较高的FOXM1表达水平与较差的生存预后相关。FOXM1的表达与糖酵解相关基因SLC2A1和LDHA呈正相关,从头脂肪生成相关基因ACACA和FASN,MYCFOXM1似乎调节AKT/mTOR信号,c-Myc的表达,与糖酵解和脂肪酸生物合成有关的蛋白质,以及HT29和HCT116细胞的细胞外酸化率。总之,FOXM1在糖酵解中起调节作用,脂肪酸生物合成,和细胞能量消耗,从而影响CRC细胞生长和患者预后。
    Metabolic reprogramming is recognized as a hallmark of cancer, enabling cancer cells to acquire essential biomolecules for cell growth, often characterized by upregulated glycolysis and/or fatty acid synthesis-related genes. The transcription factor forkhead box M1 (FOXM1) has been implicated in various cancers, contributing significantly to their development, including colorectal cancer (CRC), a major global health concern. Despite FOXM1\'s established role in cancer, its specific involvement in the Warburg effect and fatty acid biosynthesis in CRC remains unclear. We analyzed The Cancer Genome Atlas (TCGA) Colonic Adenocarcinoma and Rectal Adenocarcinoma (COADREAD) datasets to to derive the correlation of the expression levels between FOXM1 and multiple genes and the survival prognosis based on FOXM1 expression. Using two human CRC cell lines, HT29 and HCT116, we conducted RNAi or plasmid transfection procedures, followed by a series of assays, including RNA extraction, quantitative real-time polymerase chain reaction, Western blot analysis, cell metabolic assays, and immunofluorescence analysis. Higher expression levels of FOXM1 correlated with a poorer survival prognosis, and the expression of FOXM1 was positively correlated with glycolysis-related genes SLC2A1 and LDHA, de novo lipogenesis-related genes ACACA and FASN, and MYC. FOXM1 appeared to modulate AKT/mTOR signaling, the expression of c-Myc, proteins related to glycolysis and fatty acid biosynthesis, as well as extracellular acidification rate in HT29 and HCT116 cells. In summary, FOXM1 plays a regulatory role in glycolysis, fatty acid biosynthesis, and cellular energy consumption, thereby influencing CRC cell growth and patient prognosis.
    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
    尽管有流行病学迹象,二甲双胍在肝癌中的效用仍存在争议,对其抗癌作用机制的理解仍不完整。特别是,在葡萄糖充足和葡萄糖缺乏的环境下,它是否通过类似的机制起作用,或者这些作用是否可逆仍有待探索。该代谢组数据集从用二甲双胍或安慰剂治疗3小时至48小时的肝癌(HepG2)细胞以及二甲双胍停药后恢复的细胞收集。将细胞暴露于安慰剂或有或没有葡萄糖(5mM)补充的2.5mM二甲双胍。在处理后3、6、12、24和48小时收获细胞。在这些条件之一下处理24小时后,还收获细胞,然后逆转葡萄糖和/或二甲双胍暴露状态48小时。使用含有高香草酸作为内标的冷冻单相代谢物提取溶剂(水:乙腈:异丙醇=2:3:3)提取来自每个实验组的六个生物重复的代谢物。使用MOX试剂随后MSTFA对样品进行衍生化。使用与5977B单四极质谱仪偶联的Agilent7890B气相色谱仪进行衍生化样品的非靶向代谢组学分析。分析物通过不分流的衬里注入,并在使用超纯氦气作为载气的HP-5MS超惰性柱上分离。峰对齐,注释,使用AgilentMassHunter定量分析软件进行整合。使用MetaboAnalyst5.0进行多变量分析。进行这些实验是为了揭示响应二甲双胍治疗的细胞代谢组的纵向进化,它的葡萄糖依赖性,以及检查这些变化的可逆性。该数据集可以帮助鉴定参与二甲双胍抗癌作用的葡萄糖非依赖性途径。该数据集可用于设计实验,以开发与二甲双胍协同作用的新型治疗组合,以削弱癌细胞的代谢适应性。它还可以帮助开发实验来测试二甲双胍戒断对肝癌的影响。
    Despite epidemiological indications, utility of metformin in liver cancer remains debated and the understanding of the mechanism underlying its anti-cancer effects remains incomplete. Particularly, whether it operates via similar mechanism under glucose-sufficient and glucose- deficient environments or whether these effects are reversible remains unexplored. This metabolomic dataset was collected from liver cancer (HepG2) cells treated with metformin or placebo over a period of 3 h to 48 h as well as from cells recovering after metformin withdrawal. Cells were exposed to placebo or 2.5 mM metformin with or without glucose (5 mM) supplementation. The cells were harvested at 3, 6, 12, 24, and 48 h post-treatment. Cells were also harvested after 24 h of treatment under one of these conditions followed by reversal of glucose and/or metformin exposure status for 48 h. Metabolites from six biological replicates of each experimental group were extracted using chilled monophasic metabolite extraction solvent (Water: Acetonitrile: Isopropanol= 2:3:3) containing homovanillic acid as an internal standard. Samples were derivatized using MOX reagent followed by MSTFA. Untargeted metabolomic profiling of derivatized samples were performed using an Agilent 7890B gas chromatograph coupled to a 5977B single quadrupole mass spectrometer. Analytes were injected through a splitless liner and separated on a HP-5MS ultra-inert column using ultrapure helium as the carrier gas. Peak alignment, annotation, and integration were done using Agilent MassHunter Quantitative analysis software. Multivariate analysis was performed using MetaboAnalyst 5.0. These experiments were performed to unravel the longitudinal evolution of cellular metabolome in response to metformin treatment, its glucose dependence, as well as to examine the reversibility of these changes. The dataset can help to identify glucose-independent pathways involved in anti-cancer effect of metformin. The dataset can be used to design experiments to develop novel therapeutic combinations synergistically acting with metformin to cripple the metabolic fitness of cancer cells. It can also help to develop experiments to test the effect of metformin withdrawal in liver 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
    葡萄糖对能量代谢至关重要,它的使用可以决定其他蜂窝功能,取决于细胞类型。在某些病理条件下,细胞长时间暴露于高浓度的葡萄糖中。在这项研究中,我们调查了新陈代谢,氧化应激,和在生理上低(5mm)和高(12.5mm)葡萄糖浓度的培养基中培养的人支气管上皮细胞(HBEC)中的细胞衰老途径。HBEC暴露于12.5毫米葡萄糖显示增加的葡萄糖路线朝向磷酸戊糖途径,乳酸合成,和糖原,但不是甘油三酯的合成。这些代谢变化与细胞增殖率的变化无关,氧化应激,或细胞衰老途径。由于高血糖与肺纤维化有关,我们询问HBECS是否可以激活成纤维细胞。在12.5毫米葡萄糖暴露的HBEC条件下培养的原代人肺成纤维细胞显示,COL1A1和COL1A2的基因表达增加了1.3倍,平滑肌细胞肌动蛋白的蛋白质水平增加了两倍,COL1A1的蛋白质水平增加了2.4倍。始终如一,用与炎症和纤维化相关的12.5毫米葡萄糖分泌蛋白培养的HBECs,如白细胞介素IL-1β,IL-10和IL-13,CC趋化因子配体CCL2和CCL24,并与细胞外基质重塑,例如金属蛋白酶(MMP)-1,MMP-3,MMP-9和MMP-13以及MMP的组织抑制剂(TIMP)-1和-2。这项研究表明,HBECs经历代谢重编程,并在暴露于高浓度的葡萄糖后增加促纤维化介质的分泌,它有助于理解糖尿病相关肺纤维化中邻近细胞的代谢串扰。
    Glucose is essential for energy metabolism, and its usage can determine other cellular functions, depending on the cell type. In some pathological conditions, cells are exposed to high concentrations of glucose for extended periods. In this study, we investigated metabolic, oxidative stress, and cellular senescence pathways in human bronchial epithelial cells (HBECs) cultured in media with physiologically low (5 mm) and high (12.5 mm) glucose concentrations. HBECs exposed to 12.5 mm glucose showed increased glucose routing toward the pentose phosphate pathway, lactate synthesis, and glycogen, but not triglyceride synthesis. These metabolic shifts were not associated with changes in cell proliferation rates, oxidative stress, or cellular senescence pathways. Since hyperglycemia is associated with fibrosis in the lung, we asked whether HBECS could activate fibroblasts. Primary human lung fibroblasts cultured in media conditioned by 12.5 mm glucose-exposed HBECs showed a 1.3-fold increase in the gene expression of COL1A1 and COL1A2, along with twofold increased protein levels of smooth muscle cell actin and 2.4-fold of COL1A1. Consistently, HBECs cultured with 12.5 mm glucose secreted proteins associated with inflammation and fibrosis, such as interleukins IL-1β, IL-10, and IL-13, CC chemokine ligands CCL2 and CCL24, and with extracellular matrix remodeling, such as metalloproteinases (MMP)-1, MMP-3, MMP-9, and MMP-13 and tissue inhibitors of MMPs (TIMP)-1 and -2. This study shows that HBECs undergo metabolic reprogramming and increase the secretion of profibrotic mediators following exposure to high concentrations of glucose, and it contributes to the understanding of the metabolic crosstalk of neighboring cells in diabetes-associated pulmonary fibrosis.
    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
    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)

  • 文章类型: Journal Article
    氨基酸是蛋白质的重要组成部分,细胞生存的关键能源,和支持肿瘤细胞抗性生长的关键信号分子。在肿瘤细胞中,氨基酸代谢重编程的特点是氨基酸的摄取增强以及它们的异常合成,击穿,和运输,导致免疫逃避和肿瘤细胞的恶性进展。本文综述了肿瘤细胞中氨基酸代谢的改变及其对肿瘤微环境的影响。并概述了目前氨基酸代谢的临床应用。针对氨基酸代谢的创新药物在精准和个性化癌症治疗方面具有广阔的前景。
    Amino acids are essential building blocks for proteins, crucial energy sources for cell survival, and key signaling molecules supporting the resistant growth of tumor cells. In tumor cells, amino acid metabolic reprogramming is characterized by the enhanced uptake of amino acids as well as their aberrant synthesis, breakdown, and transport, leading to immune evasion and malignant progression of tumor cells. This article reviews the altered amino acid metabolism in tumor cells and its impact on tumor microenvironment, and also provides an overview of the current clinical applications of amino acid metabolism. Innovative drugs targeting amino acid metabolism hold great promise for precision and personalized cancer therapy.
    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
    动脉粥样硬化,慢性炎症,仍然是全球死亡的主要原因,需要创新的方法来靶向促动脉粥样硬化途径。免疫代谢领域的最新进展强调了动脉粥样硬化中代谢途径与免疫细胞功能之间的关键相互作用。巨噬细胞和T细胞进行动态代谢重编程,以满足活化和分化的需求,影响斑块进展。此外,代谢中间体复杂地调节免疫细胞反应和动脉粥样硬化的发展。了解动脉粥样硬化中免疫反应的代谢控制,被称为动脉粥样硬化免疫代谢,为预防和治疗干预提供了新的途径。这篇综述阐明了动脉粥样硬化中代谢和免疫之间新兴的复杂相互作用。强调代谢酶和代谢物作为疾病发病机制和治疗靶点的关键调节因子的重要性。
    Atherosclerosis, a chronic inflammatory condition, remains a leading cause of death globally, necessitating innovative approaches to target pro-atherogenic pathways. Recent advancements in the field of immunometabolism have highlighted the crucial interplay between metabolic pathways and immune cell function in atherogenic milieus. Macrophages and T cells undergo dynamic metabolic reprogramming to meet the demands of activation and differentiation, influencing plaque progression. Furthermore, metabolic intermediates intricately regulate immune cell responses and atherosclerosis development. Understanding the metabolic control of immune responses in atherosclerosis, known as athero-immunometabolism, offers new avenues for preventive and therapeutic interventions. This review elucidates the emerging intricate interplay between metabolism and immunity in atherosclerosis, underscoring the significance of metabolic enzymes and metabolites as key regulators of disease pathogenesis and therapeutic targets.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号