Tumor metabolism

肿瘤代谢
  • 文章类型: Journal Article
    精氨酸剥夺疗法(ADT)通过减少外周血精氨酸来阻碍神经胶质瘤细胞获得营养,在各种研究中表现出巨大的功效,这表明它是一种潜在的有希望的治疗神经胶质瘤的方法。本系统综述旨在探讨ADT对胶质瘤的作用机制。基于现有研究的治疗效果,和可能的联合疗法。
    我们对PubMed,根据PRISMA指南,ScienceDirect和WebofScience数据库,寻找ADT在胶质瘤中的疗效的文章。
    我们在786个搜索结果中确定了17个研究,其中ADT治疗主要基于无精氨酸条件,精氨酸脱亚胺酶和精氨酸酶,包括三项已完成的临床试验。ADT治疗已在体内和体外显示出有希望的结果,其安全性在临床试验中得到证实。在治疗的早期阶段,胶质母细胞瘤(GBM)细胞发展应激和自噬的保护机制,最终演变成胱天蛋白酶依赖性细胞凋亡或衰老,分别。精氨酸耗竭也改变了免疫抑制微环境,例如小胶质细胞转化为促炎表型和T细胞的活化。因此,ADT疗法在存在多种机制的情况下证明了神经胶质瘤的杀伤作用。结合各种常规疗法和研究药物,如放疗,替莫唑胺(TMZ),细胞周期蛋白依赖性激酶抑制剂(CDK)抑制剂和自噬诱导剂,ADT疗法已被证明更有效。然而,由于ASS1而不是干细胞的再表达而导致的耐药现象仍有待研究。
    尽管文献研究很少,现有数据证明了精氨酸剥夺疗法对神经胶质瘤的治疗潜力,并鼓励进一步研究,特别是对其联合疗法的探索,以及我们对ADT从其他肿瘤到神经胶质瘤的作用和机制的了解。
    UNASSIGNED: Arginine deprivation therapy (ADT) hinders glioma cells\' access to nutrients by reducing peripheral blood arginine, showing great efficacy in various studies, which suggests it as a potentially promising treatment for glioma. The aim of this systematic review was to explore the mechanism of ADT for gliomas, the therapeutic effect based on existing research, and possible combination therapies.
    UNASSIGNED: We performed a systematic literature review of PubMed, ScienceDirect and Web of Science databases according to PRISMA guidelines, searching for articles on the efficacy of ADT in glioma.
    UNASSIGNED: We identified 17 studies among 786 search results, among which ADT therapy mainly based on Arginine free condition, Arginine Deiminase and Arginase, including three completed clinical trials. ADT therapy has shown promising results in vivo and in vitro, with its safety confirmed in clinical trials. In the early phase of treatment, glioblastoma (GBM) cells develop protective mechanisms of stress and autophagy, which eventually evolve into caspase dependent apoptosis or senescence, respectively. The immunosuppressive microenvironment is also altered by arginine depletion, such as the transformation of microglia into a pro-inflammatory phenotype and the activation of T-cells. Thus, ADT therapy demonstrates glioma-killing effect in the presence of a combination of mechanisms. In combination with various conventional therapies and investigational drugs such as radiotherapy, temozolomide (TMZ), cyclin-dependent kinase inhibitors (CDK) inhibitors and autophagy inducers, ADT therapy has been shown to be more effective. However, the phenomenon of drug resistance due to re-expression of ASS1 rather than stem cell remains to be investigated.
    UNASSIGNED: Despite the paucity of studies in the literature, the available data demonstrate the therapeutic potential of arginine deprivation therapy for glioma and encourage further research, especially the exploration of its combination therapies and the extrapolation of what we know about the effects and mechanisms of ADT from other tumors to glioma.
    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
    胃肠道(GI)癌症给全球带来了巨大的健康负担,强调需要更深入地了解其复杂的发病机制和治疗策略。这篇综述探讨了肿瘤内微生物群之间的相互作用,肿瘤代谢,和主要类型的胃肠道癌症(包括食道癌,胃,肝脏,胰腺,和结肠直肠癌),总结最近的研究,阐明其临床意义和未来的方向。最近的研究揭示了胃肠道肿瘤中微生物特征的改变,影响肿瘤进展,免疫反应,和治疗结果。菌群失调诱导的肿瘤代谢改变,包括糖酵解,脂肪酸代谢,和氨基酸代谢,在癌症进展和治疗抗性中起关键作用。将分子机制和潜在生物标志物整合到这种理解中进一步增强了肿瘤内微生物群组成和靶向微生物群介导的肿瘤代谢的治疗机会的预后意义。尽管取得了进步,在理解肿瘤微环境(TME)内的动态相互作用方面仍然存在挑战。未来的研究方向,包括先进的组学技术和前瞻性临床研究,为胃肠道癌症的精准肿瘤学和个性化治疗干预提供了有希望的途径。总的来说,将基于微生物群的方法和分子生物标志物整合到胃肠道癌症管理中,有望改善患者预后和生存率.
    Gastrointestinal (GI) cancers impose a substantial global health burden, highlighting the necessity for deeper understanding of their intricate pathogenesis and treatment strategies. This review explores the interplay between intratumoral microbiota, tumor metabolism, and major types of GI cancers (including esophageal, gastric, liver, pancreatic, and colorectal cancers), summarizing recent studies and elucidating their clinical implications and future directions. Recent research revealed altered microbial signatures within GI tumors, impacting tumor progression, immune responses, and treatment outcomes. Dysbiosis-induced alterations in tumor metabolism, including glycolysis, fatty acid metabolism, and amino acid metabolism, play critical roles in cancer progression and therapeutic resistance. The integration of molecular mechanisms and potential biomarkers into this understanding further enhances the prognostic significance of intratumoral microbiota composition and therapeutic opportunities targeting microbiota-mediated tumor metabolism. Despite advancements, challenges remain in understanding the dynamic interactions within the tumor microenvironment (TME). Future research directions, including advanced omics technologies and prospective clinical studies, offer promising avenues for precision oncology and personalized treatment interventions in GI cancer. Overall, integrating microbiota-based approaches and molecular biomarkers into GI cancer management holds promise for improving patient outcomes and survival.
    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
    癌症对人类健康和生命构成重大威胁。化疗,免疫疗法和化学动力学疗法(CDT)是癌症的有效治疗方法。然而,肿瘤细胞中通过谷氨酰胺进行代谢重编程的存在限制了其治疗效果.在这里,我们提出了一种有效的组装策略来合成一种新型的基于金属-多酚的多功能纳米药物(Fe-DBEF),其中包含PluronicF127稳定的铁离子交联的表没食子儿茶素没食子酸酯(EGCG)纳米颗粒,负载有GLS1抑制剂双-2-(5-苯基乙酰氨基-1,3,4-噻二唑-2-基)乙基硫醚(BPTES)和我们的研究表明,通过结合体外细胞实验,Fe-DBEF纳米药物在胰腺癌中表现出高效的抗增殖特性,人体类器官实验和KPC动物实验。值得注意的是,Fe-DBEF纳米药物可以减少肿瘤细胞中谷胱甘肽(GSH)的产生,从而降低其对ROS治疗的抗性。此外,过量的ROS产生也会加剧DOX引起的DNA损伤,协同增敏化疗和促进细胞凋亡有效治疗胰腺癌。总的来说,我们的研究结果表明,使用基于金属-多酚的多功能纳米药物抑制谷氨酰胺代谢以增加化疗/CDT的敏感性,为胰腺癌的治疗提供了一种有前景的多种治疗手段的组合.
    Cancer poses a significant threat to human health and life. Chemotherapy, immunotherapy and chemodynamic therapy (CDT) are effective treatments for cancer. However, the presence of metabolic reprogramming via glutamine in tumor cells limits their therapeutic effectiveness. Herein, we propose an effective assembly strategy to synthesize a novel metal-polyphenolic based multifunctional nanomedicine (Fe-DBEF) containing Pluronic F127 stable ferric ion crosslinked epigallocatechin gallate (EGCG) nanoparticles loaded with GLS1 inhibitor bis-2-(5-phenylacetamino-1,3,4-thiadiazole-2-yl) ethyl sulfide (BPTES) and chemotherapy drug doxorubicin (DOX). Our study demonstrates that Fe-DBEF nanomedicine exhibits high efficiency anti-proliferation properties in pancreatic cancer through a combination of in vitro cell experiments, human organoid experiments and KPC animal experiments. Notably, Fe-DBEF nanomedicine can reduce the production of glutathione (GSH) in tumor cells, thereby reducing their resistance to ROS therapy. Additionally, excessive ROS production also aggravates DNA damage caused by DOX, synergistically sensitizing chemotherapy and promoting apoptosis for efficient treatment of pancreatic cancer. Overall, our findings suggest that inhibiting glutamine metabolism to increase the sensitivity of chemotherapy/CDT using metal-polyphenolic based multifunctional nanomedicine provides a promising combination of multiple therapeutic means for treating pancreatic cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肿瘤微环境表现出巨大的免疫表型异质性,基于肿瘤内免疫细胞的丰度,在传统的免疫热/冷肿瘤分类中得到了利用。通过结合空间免疫背景,肿瘤免疫表型进一步细化为免疫炎症,免疫排除,和免疫沙漠。然而,这些不同免疫表型的潜在机制尚待全面阐明。在这次审查中,我们从肿瘤细胞的角度讨论了肿瘤细胞和肿瘤微环境如何共同相互作用以塑造免疫景观,免疫细胞,细胞外基质,和癌症代谢,我们根据不同的免疫表型总结了个性化精准医疗的潜在治疗选择。
    The tumor microenvironment demonstrates great immunophenotypic heterogeneity, which has been leveraged in traditional immune-hot/cold tumor categorization based on the abundance of intra-tumoral immune cells. By incorporating the spatial immune contexture, the tumor immunophenotype was further elaborated into immune-inflamed, immune-excluded, and immune-desert. However, the mechanisms underlying these different immune phenotypes are yet to be comprehensively elucidated. In this review, we discuss how tumor cells and the tumor microenvironment interact collectively to shape the immune landscape from the perspectives of tumor cells, immune cells, the extracellular matrix, and cancer metabolism, and we summarize potential therapeutic options according to distinct immunophenotypes for personalized precision medicine.
    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
    非利尿钠依赖性谷氨酸/胱氨酸反向转运系统Xc-由两个蛋白质亚基组成,SLC7A11和SLC3A2,其中SLC7A11作为负责胱氨酸摄取和谷胱甘肽生物合成的主要功能成分。SLC7A11通过调节氧化还原稳态参与肿瘤的发展,氨基酸代谢,调节免疫功能,以及细胞程序性死亡的诱导,在与肿瘤发生相关的其他过程中。在本文中,本文综述了SLC7A11的结构和生物学功能,并讨论了其在肿瘤治疗中的潜在作用,为肿瘤的精准化、个性化治疗提供了新的方向。
    The non-natriuretic-dependent glutamate/cystine inverse transporter-system Xc- is composed of two protein subunits, SLC7A11 and SLC3A2, with SLC7A11 serving as the primary functional component responsible for cystine uptake and glutathione biosynthesis. SLC7A11 is implicated in tumor development through its regulation of redox homeostasis, amino acid metabolism, modulation of immune function, and induction of programmed cell death, among other processes relevant to tumorigenesis. In this paper, we summarize the structure and biological functions of SLC7A11, and discuss its potential role in tumor therapy, which provides a new direction for precision and personalized treatment of tumors.
    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
    肉碱棕榈酰转移酶1C(CPT1C)是一种通过调节线粒体功能和脂质代谢来调节肿瘤细胞增殖和代谢的酶。缺氧,通常在实体瘤中观察到,通过调节肿瘤细胞的代谢重编程促进胰腺癌的增殖和进展。到目前为止,CPT1C对缺氧肿瘤细胞的代谢调节和CPT1C的上游机制仍然知之甚少。阴阳1(YY1)是胰腺肿瘤发生的关键癌基因,是参与多种代谢过程的转录因子。本研究旨在阐明YY1和CPT1C在低氧条件下的关系,探讨其在低氧诱导的肿瘤细胞增殖和代谢改变中的作用。结果表明,低氧条件下PANC-1细胞的增殖和代谢增强,细胞生长增加证明了这一点,细胞ATP水平,线粒体膜电位的上调,和降低脂质含量。有趣的是,敲除YY1或CPT1C抑制低氧诱导的快速细胞增殖和活跃的细胞代谢。重要的是,第一次,我们报道了YY1直接激活CPT1C的转录,并阐明了CPT1C是YY1的新靶基因。此外,通过用YY1siRNA转染CRISPR/Cas9-CPT1C敲除的PANC-1细胞,发现YY1和CPT1C协同调节低氧细胞的增殖和代谢.一起来看,提示YY1-CPT1C轴可能成为干预胰腺癌增殖和代谢的新靶点。
    Carnitine palmitoyltransferase 1C (CPT1C) is an enzyme that regulates tumor cell proliferation and metabolism by modulating mitochondrial function and lipid metabolism. Hypoxia, commonly observed in solid tumors, promotes the proliferation and progression of pancreatic cancer by regulating the metabolic reprogramming of tumor cells. So far, the metabolic regulation of hypoxic tumor cells by CPT1C and the upstream mechanisms of CPT1C remain poorly understood. Yin Yang 1 (YY1) is a crucial oncogene for pancreatic tumorigenesis and acts as a transcription factor that is involved in multiple metabolic processes. This study aimed to elucidate the relationship between YY1 and CPT1C under hypoxic conditions and explore their roles in hypoxia-induced proliferation and metabolic alterations of tumor cells. The results showed enhancements in the proliferation and metabolism of PANC-1 cells under hypoxia, as evidenced by increased cell growth, cellular ATP levels, up-regulation of mitochondrial membrane potential, and decreased lipid content. Interestingly, knockdown of YY1 or CPT1C inhibited hypoxia-induced rapid cell proliferation and vigorous cell metabolism. Importantly, for the first time, we reported that YY1 directly activated the transcription of CPT1C and clarified that CPT1C was a novel target gene of YY1. Moreover, the YY1 and CPT1C were found to synergistically regulate the proliferation and metabolism of hypoxic cells through transfection with YY1 siRNA to CRISPR/Cas9-CPT1C knockout PANC-1 cells. Taken together, these results indicated that the YY1-CPT1C axis could be a new target for the intervention of pancreatic cancer proliferation and 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
    顺铂(CDDP)是标准的非小细胞肺癌(NSCLC)化疗,但是它的功效受到阻力的阻碍,部分原因是Warburg效应。这项研究调查了甲状腺激素如何增强Warburg效应,肺癌对顺铂的敏感性增加。根据甲状腺激素水平分析晚期非小细胞肺癌患者的临床资料,将患者分为高组和低组。细胞实验涉及对照,10uMCDDP,10uMCDDP+0.1uMT3和10uMCDDP+0.1uMT4类别。在A549和PC9肺癌细胞中测量参数,包括扩散,凋亡,线粒体膜电位,ROS生产,糖酵解酶活性,乳酸水平,和ATP含量。使用qPCR和蛋白质印迹评估基因和蛋白质表达。分析显示,较高的FT3水平与化疗前无进展生存期延长相关(中位PFS:高FT3组=12.67个月,低FT3组=7.03个月,p=0.01)。细胞实验表明,甲状腺激素增加肺癌细胞对顺铂的敏感性,抑制增殖和增强功效。机制涉及甲状腺激素和顺铂共同下调MSI1/AKT/GLUT1表达,减少乳酸和糖酵解。这种Warburg效应逆转提高了ATP水平,提升ROS,并减少MMP,增强顺铂在A549和PC9细胞中的有效性。总之,晚期NSCLC患者游离T3水平升高与顺铂化疗组无进展生存期延长相关.细胞实验表明,甲状腺激素通过逆转Warburg效应增强肺癌细胞对顺铂的敏感性,为改善治疗结果提供机制基础。
    Cisplatin (CDDP) is a standard non-small cell lung cancer (NSCLC) chemotherapy, but its efficacy is hampered by resistance, partly due to the Warburg effect. This study investigates how thyroid hormones enhance the Warburg effect, increasing sensitivity to cisplatin in lung cancer. Clinical data from advanced NSCLC patients were analyzed based on thyroid hormone levels, categorizing patients into high and low groups. Cellular experiments involved Control, 10uM CDDP, 10uM CDDP + 0.1uM T3, and 10uM CDDP + 0.1uM T4 categories. Parameters were measured in A549 and PC9 lung cancer cells, including proliferation, apoptosis, mitochondrial membrane potential, ROS production, glycolysis enzyme activity, lactic acid level, and ATP content. Gene and protein expressions were assessed using qPCR and Western Blot. Analysis revealed higher FT3 levels correlated with prolonged progression-free survival before chemotherapy (median PFS: high FT3 group = 12.67 months, low FT3 group = 7.03 months, p = 0.01). Cellular experiments demonstrated that thyroid hormones increase lung cancer cell sensitivity to cisplatin, inhibiting proliferation and enhancing efficacy. The mechanism involves thyroid hormones and cisplatin jointly down-regulating MSI1/AKT/GLUT1 expression, reducing lactic acid and glycolysis. This Warburg effect reversal boosts ATP levels, elevates ROS, and decreases MMP, enhancing cisplatin effectiveness in A549 and PC9 cells. In conclusion, elevated free T3 levels in advanced NSCLC patients correlate with prolonged progression-free survival under cisplatin chemotherapy. Cellular experiments reveal that thyroid hormones enhance lung cancer cell sensitivity to cisplatin by reversing the Warburg effect, providing a mechanistic basis for improved therapeutic outcomes.
    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
    肿瘤采用各种策略来逃避免疫监视。中枢神经体系(CNS)具有克制免疫反响的多重特征。肿瘤和中枢神经系统是否具有相似的免疫抑制方案是难以捉摸的。这里,我们分析了接受曲妥珠单抗和抗PD-L1抗体的HER2+乳腺癌患者肿瘤的多组学数据,发现富含CNS的N-乙酰转移酶8样(NAT8L)及其代谢物N-乙酰天冬氨酸(NAA)在耐药肿瘤中过表达.在CNS中,NAA在脑炎症期间释放。NAT8L通过经由NAA抑制自然杀伤(NK)细胞和CD8+T细胞的细胞毒性来减轻脑部炎症并损害抗肿瘤免疫。NAA通过促进PCAF诱导的层粘连蛋白A-K542的乙酰化来破坏免疫突触的形成,从而抑制层粘连蛋白A和SUN2之间的整合并削弱裂解颗粒的极化。我们发现肿瘤细胞模拟CNS的抗炎机制以逃避抗肿瘤免疫,NAT8L是增强抗癌剂功效的潜在靶标。
    Tumors employ various strategies to evade immune surveillance. Central nervous system (CNS) has multiple features to restrain immune response. Whether tumors and CNS share similar programs of immunosuppression is elusive. Here, we analyze multi-omics data of tumors from HER2+ breast cancer patients receiving trastuzumab and anti-PD-L1 antibody and find that CNS-enriched N-acetyltransferase 8-like (NAT8L) and its metabolite N-acetylaspartate (NAA) are overexpressed in resistant tumors. In CNS, NAA is released during brain inflammation. NAT8L attenuates brain inflammation and impairs anti-tumor immunity by inhibiting cytotoxicity of natural killer (NK) cells and CD8+ T cells via NAA. NAA disrupts the formation of immunological synapse by promoting PCAF-induced acetylation of lamin A-K542, which inhibits the integration between lamin A and SUN2 and impairs polarization of lytic granules. We uncover that tumor cells mimic the anti-inflammatory mechanism of CNS to evade anti-tumor immunity and NAT8L is a potential target to enhance efficacy of anti-cancer agents.
    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
    与癌症相关的RNA甲基化改变的发现有望将其用作癌症诊断的潜在生物标志物。预后,和预测。已发现RNA甲基化影响肿瘤的免疫微环境,但是甲基化相关基因(MRGs)的特定作用,特别是在乳腺癌(BC),全球女性中最常见的癌症,肿瘤内的微环境仍然未知。在这项研究中,我们从TCGA和GEO数据库获得数据,以研究MRGs在BC基因组和转录域中的表达模式.通过分析数据,我们确定了与临床病理特征相关的两个不同的遗传分组,预后,TME细胞浸润程度,以及患者中MRG的其他异常。随后,建立了MRG模型来预测总生存期(OS),并在BC患者中评估了其准确性.此外,创建了高度精确的列线图,以增强MRG模型的实际可用性.在低风险人群中,我们观察到较低的TBM值和较高的TIDE评分.我们进一步探讨了MRGs如何影响患者的预后,临床显著特征,对治疗的反应,还有TME.这些风险特征有可能改善BC患者的治疗策略,并可应用于未来的临床环境。此外,它们也可用于确定这些患者的预后和生物学特征。
    The discovery of RNA methylation alterations associated with cancer holds promise for their utilization as potential biomarkers in cancer diagnosis, prognosis, and prediction. RNA methylation has been found to impact the immunological microenvironment of tumors, but the specific role of methylation-related genes (MRGs), particularly in breast cancer (BC), the most common cancer among women globally, within the tumor microenvironment remains unknown. In this study, we obtained data from TCGA and GEO databases to investigate the expression patterns of MRGs in both genomic and transcriptional domains in BC. By analyzing the data, we identified two distinct genetic groupings that were correlated with clinicopathological characteristics, prognosis, degree of TME cell infiltration, and other abnormalities in MRGs among patients. Subsequently, an MRG model was developed to predict overall survival (OS) and its accuracy was evaluated in BC patients. Additionally, a highly precise nomogram was created to enhance the practical usability of the MRG model. In low-risk groups, we observed lower TBM values and higher TIDE scores. We further explored how MRGs influence a patient\'s prognosis, clinically significant characteristics, response to therapy, and the TME. These risk signatures have the potential to improve treatment strategies for BC patients and could be applied in future clinical settings. Moreover, they may also be utilized to determine prognosis and biological features in these patients.
    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
    肿瘤细胞的能量需求极高,因此肿瘤细胞代谢的核心是能量代谢的管理。细胞中的嘌呤核苷酸合成途径使用嘌呤小体作为必需的空间结构复合物。除了在肿瘤的出现和生长中起着至关重要的调节作用之外,它有助于嘌呤核苷酸的合成和代谢。本文首先讨论了嘌呤在肿瘤细胞中代谢的重要性。然后回顾了嘌呤体作为前瞻性治疗靶标的作用,以及一系列在肿瘤代谢调节中起作用的信号通路。彻底了解嘌呤在控制肿瘤代谢中的功能可以为创建创新的癌症治疗方法提供新的建议。
    The core of tumor cell metabolism is the management of energy metabolism due to the extremely high energy requirements of tumor cells. The purine nucleotide synthesis pathway in cells uses the purinosomes as an essential spatial structural complex. In addition to serving a crucial regulatory role in the emergence and growth of tumors, it contributes to the synthesis and metabolism of purine nucleotides. The significance of purine metabolism in tumor cells is initially addressed in this current article. The role of purinosomes as prospective therapeutic targets is then reviewed, along with a list of the signaling pathways that play in the regulation of tumor metabolism. A thorough comprehension of the function of purinosomes in the control of tumor metabolism can generate fresh suggestions for the creation of innovative cancer treatment methods.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号