cancer metabolism

癌症代谢
  • 文章类型: Journal Article
    乳酸脱氢酶A(LDHA)在许多肿瘤细胞中高表达,并在葡萄糖途径中增进丙酮酸向乳酸的转化,为肿瘤细胞的快速增殖提供能量和合成前体。因此,抑制LDHA已成为广泛关注的肿瘤治疗策略。然而,高效低毒的LDHA小分子抑制剂的研发仍面临挑战。为了发现潜在的LDHA抑制剂,基于分子对接技术的虚拟筛选来自超过260,000种化合物的Specs数据库和超过1,000种化合物的Chemdiv-smart数据库.通过分子动力学(MD)模拟研究,我们确定了12种潜在的LDHA抑制剂,所有这些都可以稳定地结合人LDHA蛋白并与其活性中心残基形成多重相互作用。为了验证这些化合物的抑制活性,我们建立了酶活性测定系统,并测定了它们对重组人LDHA的抑制作用。结果表明,化合物6可以剂量依赖的方式抑制LDHA对丙酮酸的催化作用,EC50值为14.54±0.83µM。进一步的体外实验表明,化合物6能显著抑制胰腺癌细胞、肺癌细胞等多种肿瘤细胞系的增殖,降低细胞内乳酸含量并增加细胞内活性氧(ROS)水平。总之,通过虚拟筛选和体外验证,我们发现化合物6是LDHA的小分子抑制剂,为LDHA相关靶向抗肿瘤药物的研发提供了良好的先导化合物。
    Lactate dehydrogenase A (LDHA) is highly expressed in many tumor cells and promotes the conversion of pyruvate to lactic acid in the glucose pathway, providing energy and synthetic precursors for rapid proliferation of tumor cells. Therefore, inhibition of LDHA has become a widely concerned tumor treatment strategy. However, the research and development of highly efficient and low toxic LDHA small molecule inhibitors still faces challenges. To discover potential inhibitors against LDHA, virtual screening based on molecular docking techniques was performed from Specs database of more than 260,000 compounds and Chemdiv-smart database of more than 1,000 compounds. Through molecular dynamics (MD) simulation studies, we identified 12 potential LDHA inhibitors, all of which can stably bind to human LDHA protein and form multiple interactions with its active central residues. In order to verify the inhibitory activities of these compounds, we established an enzyme activity assay system and measured their inhibitory effects on recombinant human LDHA. The results showed that Compound 6 could inhibit the catalytic effect of LDHA on pyruvate in a dose-dependent manner with an EC50 value of 14.54 ± 0.83 µM. Further in vitro experiments showed that Compound 6 could significantly inhibit the proliferation of various tumor cell lines such as pancreatic cancer cells and lung cancer cells, reduce intracellular lactic acid content and increase intracellular reactive oxygen species (ROS) level. In summary, through virtual screening and in vitro validation, we found that Compound 6 is a small molecule inhibitor for LDHA, providing a good lead compound for the research and development of LDHA related targeted anti-tumor drugs.
    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
    醛缩酶,特别是ALDOA,ALDOB,和ALDOC,在癌症的发展和进展中起着至关重要的作用。虽然醛缩酶家族主要以参与糖酵解途径而闻名,这些酶还通过不同的信号通路,如Wnt/β-catenin,EGFR/MAPK,Akt,和HIF-1α。近年来,这引起了越来越多的关注,并揭示了这种酶的其他方面。靶向醛缩酶的潜在治疗策略包括使用siRNA,抑制剂如萘酚AS-E磷酸酯和TX-2098,以及天然化合物如HDPS-4II和L-肌肽。此外,来自ALDOA的抗癌肽,像P04一样,可能会增加癌细胞对化疗的敏感性。醛缩酶还通过不同的方法影响癌症的耐药性,使它们成为良好的治疗目标。在这次审查中,我们广泛探索醛缩酶在各种癌症增殖中的作用,入侵,迁移,和耐药性;我们还显着探索考虑醛缩酶功能的可能治疗方法。
    Aldolase enzymes, particularly ALDOA, ALDOB, and ALDOC, play a crucial role in the development and progression of cancer. While the aldolase family is mainly known for its involvement in the glycolysis pathway, these enzymes also have various pathological and physiological functions through distinct signaling pathways such as Wnt/β-catenin, EGFR/MAPK, Akt, and HIF-1α. This has garnered increased attention in recent years and shed light on other sides of this enzyme. Potential therapeutic strategies targeting aldolases include using siRNA, inhibitors like naphthol AS-E phosphate and TX-2098, and natural compounds such as HDPS-4II and L-carnosine. Additionally, anticancer peptides derived from ALDOA, like P04, can potentially increase cancer cells\' sensitivity to chemotherapy. Aldolases also affect cancer drug resistance by different approaches, making them good therapeutic targets. In this review, we extensively explore the role of aldolase enzymes in various types of cancers in proliferation, invasion, migration, and drug resistance; we also significantly explore the possible treatment considering aldolase function.
    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
    作为一种具有高糖酵解活性的实体瘤,肝细胞癌(HCC)产生过量的乳酸和增加细胞外酸度,从而形成独特的免疫抑制微环境。L-乳酸脱氢酶(LDH)和单羧酸转运体(MCT)在糖酵解中起着非常重要的作用。LDH是乳酸(LA)生产的关键酶,MCT负责LA的细胞进出口。两者的协同作用促进了细胞外酸性微环境的形成。在肝癌的酸性微环境中,洛杉矶不仅可以促进扩散,生存,肿瘤细胞的运输和血管生成,但对免疫细胞也有强烈的影响,最终导致抑制性免疫微环境。本文综述了LA在HCC中的作用。尤其是它对免疫细胞的影响,总结了LDH和MCT相关药物的研究进展,并强调了针对乳酸联合肝癌的免疫治疗的潜力。
    As a solid tumor with high glycolytic activity, hepatocellular carcinoma (HCC) produces excess lactic acid and increases extracellular acidity, thus forming a unique immunosuppressive microenvironment. L-lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs) play a very important role in glycolysis. LDH is the key enzyme for lactic acid (LA) production, and MCT is responsible for the cellular import and export of LA. The synergistic effect of the two promotes the formation of an extracellular acidic microenvironment. In the acidic microenvironment of HCC, LA can not only promote the proliferation, survival, transport and angiogenesis of tumor cells but also have a strong impact on immune cells, ultimately leading to an inhibitory immune microenvironment. This article reviews the role of LA in HCC, especially its effect on immune cells, summarizes the progress of LDH and MCT-related drugs, and highlights the potential of immunotherapy targeting lactate combined with HCC.
    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
    乳酸是体内产生的糖酵解的代谢产物,它的产生被认为是癌细胞逃避免疫监视的一种机制。免疫逃避和代谢变化是癌症的基本标志。虽然乳酸一直被认为是废物,它现在被公认为是一种多功能的小分子化学物质,在肿瘤微环境(TME)中起着重要作用,乳酸产量增加与人类恶性肿瘤的发展有关。肝癌的代谢转向糖酵解,这增强了肿瘤细胞用来生产蛋白质的代谢化合物的产生,脂质,和核苷酸,使它们能够保持高的增殖率并建立TME。由于肿瘤细胞中厌氧糖酵解速率产生的乳酸的免疫抑制活性,肝癌中代谢活性的失调可能会损害抗肿瘤反应。这篇综述主要探讨了乳酸与TME之间的联系;评估了乳酸在TME发生中的作用,转移,预后,和肝癌的治疗。此外,它研究了相关的途径作为肝癌治疗的潜在目标。
    文献检索在PubMed中进行,WebofScience,和谷歌学者,最近一篇文章的发表日期为2024年1月。在通过标题和摘要消除重复的文章和不太相关的文章之后,我们选择了113篇文章进行这篇评论。我们将参考文献分为两类。一种是将含量分为与乳酸相关的,肝癌相关和肿瘤代谢相关。另一种是对文章类型进行分类,分为评论,研究文章和临床试验。此外,我们查阅了相关文章的参考清单,以确保报道全面和公正。
    乳酸与TME之间的联系最近已成为一个备受关注的研究领域,和许多相关的文章已经发表在这一领域。这篇综述的主要发现是总结乳酸与TME之间的已证实的联系及其对肝癌TME的可能影响。并分析乳酸在肝癌治疗及预后预测中的潜力。
    乳酸可能是开发未来肝癌治疗新方法的关键。经典疗法与分子靶向药物结合的相关研究可能提供更具选择性地调节免疫细胞活性的创新药物。
    UNASSIGNED: Lactic acid is a metabolite of glycolysis produced in the body, and its production is thought to be a mechanism by which cancer cells evade immune surveillance. Immune evasion and metabolic changes are well established as basic hallmarks of cancer. Although lactate has long been considered a waste product, it is now generally recognized to be a versatile small-molecule chemical that plays an important part in the tumor microenvironment (TME), with increased lactate production linked to the development of human malignancies. Metabolism in liver cancer is redirected toward glycolysis, which enhances the production of metabolic compounds used by tumor cells to produce proteins, lipids, and nucleotides, enabling them to maintain high proliferation rates and to establish the TME. Dysregulation of metabolic activity in liver cancer may impair antitumor responses owing to the immunosuppressive activity of the lactate produced by anaerobic glycolytic rates in tumor cells. This review primarily explores the link connection between lactic acid and the TME; evaluates the role of lactic acid in the occurrence, metastasis, prognosis, and treatment of liver cancer. Additionally, it investigates the associated pathways as potential targets for liver cancer treatment.
    UNASSIGNED: Literature searches were conducted in PubMed, Web of Science, and Google Scholar, with the publication date of the most recent article included being January 2024. After eliminating duplicate articles and less relevant articles through titles and abstracts, we selected 113 articles for this review. We categorized references into two categories. One is to classify the content into lactate-related, liver cancer-related and tumor metabolism-related. The other is to classify the article types, which are divided into reviews, research articles and clinical trials. Additionally, we consulted the reference lists of the relevant articles to ensure coverage was comprehensive and unbiased.
    UNASSIGNED: The connection between lactic acid and the TME has recently become an area of intense research interest, and many related articles have been published in this field. The main finding of this review is to summarize the proven link between lactate and the TME and its possible impact on the TME of liver cancer. And analyzed the potential of lactate in liver cancer treatment and prognosis prediction.
    UNASSIGNED: Lactate may be key to developing novel approaches in the future treatment of liver cancer. Related research on the combination of classic therapies and molecular targeted drugs may provide innovative medicines that more selectively regulate immune cell activity.
    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
    胃癌,全球第五大流行癌症,通常诊断为晚期,治疗选择有限。检查肿瘤微环境(TME)及其代谢重编程可以为更好的诊断和治疗提供见解。本研究使用批量和单细胞RNA测序数据调查了TME因子与胃癌代谢活性之间的联系。我们通过分析与TME和代谢相关的81个预后基因的不同表达模式,确定了胃癌中的两个分子亚型。表现出显著的蛋白质水平相互作用。高风险亚型的基质含量增加,成纤维细胞和M2巨噬细胞浸润,升高的糖胺聚糖/鞘糖脂生物合成,和脂肪代谢,以及先进的临床病理特征。它还表现出低突变率和微卫星不稳定性,将其与间充质表型相关联。相比之下,低危组肿瘤含量较高,蛋白质和糖代谢上调.我们确定了代表这些特征的15个基因预后特征,包括CPVL,KYNU,CD36和GPX3与M2巨噬细胞密切相关,通过单细胞分析和内部队列验证。尽管对免疫疗法有抗药性,高危人群对针对IGF-1R(BMS-754807)和PI3K-mTOR通路(AZD8186,AZD8055)的分子靶向药物表现出敏感性.我们通过实验验证了这些有前途的药物对MKN45和MKN28胃细胞的抑制作用。这项研究揭示了胃癌中TME和代谢途径之间复杂的相互作用,提供增强诊断的潜力,患者分层,个性化治疗。了解每个亚型的分子特征丰富了我们对胃癌异质性和潜在治疗靶点的理解。
    Gastric cancer, the fifth most prevalent cancer worldwide, is often diagnosed in advanced stages with limited treatment options. Examining the tumor microenvironment (TME) and its metabolic reprogramming can provide insights for better diagnosis and treatment. This study investigates the link between TME factors and metabolic activity in gastric cancer using bulk and single-cell RNA-sequencing data. We identified two molecular subtypes in gastric cancer by analyzing the distinct expression patterns of 81 prognostic genes related to the TME and metabolism, which exhibited significant protein-level interactions. The high-risk subtype had increased stromal content, fibroblast and M2 macrophage infiltration, elevated glycosaminoglycans/glycosphingolipids biosynthesis, and fat metabolism, along with advanced clinicopathological features. It also exhibited low mutation rates and microsatellite instability, associating it with the mesenchymal phenotype. In contrast, the low-risk group showed higher tumor content and upregulated protein and sugar metabolism. We identified a 15-gene prognostic signature representing these characteristics, including CPVL, KYNU, CD36, and GPX3, strongly correlated with M2 macrophages, validated through single-cell analysis and an internal cohort. Despite resistance to immunotherapy, the high-risk group showed sensitivity to molecular targeted agents directed at IGF-1R (BMS-754807) and the PI3K-mTOR pathways (AZD8186, AZD8055). We experimentally validated these promising drugs for their inhibitory effects on MKN45 and MKN28 gastric cells. This study unveils the intricate interplay between TME and metabolic pathways in gastric cancer, offering potential for enhanced diagnosis, patient stratification, and personalized treatment. Understanding molecular features in each subtype enriches our comprehension of gastric cancer heterogeneity and potential therapeutic targets.
    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
    癌症代谢异常导致缺氧和免疫抑制肿瘤微环境(TME),这限制了光动力疗法(PDT)的抗肿瘤功效。在这里,我们报告了具有锚定的3-溴丙酮酸(BrP)的光敏纳米级金属有机层(MOL),BrP@MOL,作为代谢重编程剂,以增强PDT和抗肿瘤免疫力。BrP@MOL抑制线粒体呼吸和糖酵解以氧化肿瘤并减少乳酸产生。这种代谢重编程增强了PDT过程中活性氧的产生,并重塑了免疫抑制性TME以增强抗肿瘤免疫力。BrP@MOL介导的PDT抑制肿瘤生长>90%,治愈率为40%,拒绝肿瘤再攻击,并防止肺转移。与免疫检查点阻断的进一步组合以>98%的肿瘤抑制和80%的治愈率有效地消退了肿瘤。
    Abnormal cancer metabolism causes hypoxic and immunosuppressive tumor microenvironment (TME), which limits the antitumor efficacy of photodynamic therapy (PDT). Herein, we report a photosensitizing nanoscale metal-organic layer (MOL) with anchored 3-bromopyruvate (BrP), BrP@MOL, as a metabolic reprogramming agent to enhance PDT and antitumor immunity. BrP@MOL inhibited mitochondrial respiration and glycolysis to oxygenate tumors and reduce lactate production. This metabolic reprogramming enhanced reactive oxygen species generation during PDT and reshaped the immunosuppressive TME to enhance antitumor immunity. BrP@MOL-mediated PDT inhibited tumor growth by >90 % with 40 % of mice being tumor-free, rejected tumor re-challenge, and prevented lung metastasis. Further combination with immune checkpoint blockade potently regressed the tumors with >98 % tumor inhibition and 80 % of mice being tumor-free.
    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
    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)

  • 文章类型: Journal Article
    IDH1的突变通常在各种癌症中观察到,导致α-KG转换为2-HG。2-HG水平升高会破坏组蛋白和DNA去甲基化过程,促进肿瘤发展。因此,对开发靶向突变酶的小分子抑制剂有很大的兴趣。在这里,我们报告了使用天然产物库的基于结构的高通量虚拟筛选策略,其次是命中领先优化。通过这个过程,我们发现了一种有效的化合物,名为11s,对IDH1R132H和IDH1R132C表现出显著的抑制作用,IC50值分别为124.4和95.7nM,分别。此外,11s有效地减少了2-HG的形成,在U87R132H细胞中EC50值为182nM,和84nM在HT-1080细胞中。此外,11s显着降低U87R132H和HT-1080细胞增殖,GC50值为3.48和1.38μM,分别。PK-PD实验进一步证实,化合物11s显着降低HT-1080异种移植小鼠模型中2-HG的形成,导致肿瘤生长的显著抑制,而体重没有明显下降。
    Mutations in IDH1 are commonly observed across various cancers, causing the conversion of α-KG to 2-HG. Elevated levels of 2-HG disrupt histone and DNA demethylation processes, promoting tumor development. Consequently, there is substantial interest in developing small molecule inhibitors targeting the mutant enzymes. Herein, we report a structure-based high-throughput virtual screening strategy using a natural products library, followed by hit-to-lead optimization. Through this process, we discover a potent compound, named 11s, which exhibited significant inhibition to IDH1 R132H and IDH1 R132C with IC50 values of 124.4 and 95.7 nM, respectively. Furthermore, 11s effectively reduced 2-HG formation, with EC50 values of 182 nM in U87 R132H cell, and 84 nM in HT-1080 cell. In addition, 11s significantly reduced U87 R132H and HT-1080 cell proliferation with GC50 values of 3.48 and 1.38 μM, respectively. PK-PD experiments further confirmed that compound 11s significantly decreased 2-HG formation in an HT-1080 xenograft mouse model, resulting in notable suppression of tumor growth without apparent loss in body weight.
    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
    据报道,KDM6A(赖氨酸脱甲基酶6A)在结直肠癌中经历失活突变,但其在结直肠癌进展中的功能尚未使用结直肠癌动物模型进行评估。在这项研究中,我们发现敲除小鼠肠上皮中KDM6A的表达会增加绒毛和隐窝的长度,促进AOM(偶氮甲烷)/DSS(葡聚糖硫酸钠盐)诱导的结直肠癌的发展。另一方面,敲低KDM6A的表达促进结直肠癌细胞的生长。在分子机制研究中,我们发现KDM6A与HIF-1α相互作用;敲低KDM6A促进HIF-1α与LDHA启动子的结合,从而促进LDHA表达和乳酸产生,增强糖酵解。敲低LDHA逆转了由KDM6A表达损失引起的恶性表型。总之,这项使用动物模型的研究表明,KDM6A丢失通过重编程大肠癌细胞的代谢促进大肠癌的进展,提示恢复KDM6A的功能可能是结直肠癌治疗的策略之一.
    KDM6A (lysine demethylase 6A) has been reported to undergo inactivating mutations in colorectal cancer, but its function in the progression of colorectal cancer has not been evaluated using animal models of colorectal cancer. In this study, we found that knocking out KDM6A expression in mouse intestinal epithelium increased the length of villus and crypt, promoting the development of AOM (azoxymethane)/DSS (dextran sulfate sodium salt)-induced colorectal cancer. On the other hand, knocking down KDM6A expression promoted the growth of colorectal cancer cells. In molecular mechanism studies, we found that KDM6A interacts with HIF-1α; knocking down KDM6A promotes the binding of HIF-1α to the LDHA promoter, thereby promoting LDHA expression and lactate production, enhancing glycolysis. Knocking down LDHA reversed the malignant phenotype caused by KDM6A expression loss. In summary, this study using animal models revealed that KDM6A loss promotes the progression of colorectal cancer through reprogramming the metabolism of the colorectal cancer cells, suggesting that restoring the function of KDM6A is likely to be one of the strategies for colorectal cancer treatment.
    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
    PGC1α,线粒体生物学的核心角色,在癌细胞中看到的代谢变化中具有复杂的作用。虽然它的失调在主要癌症中很常见,其影响各不相同。在某些情况下,下调促进有氧糖酵解和进展,而在其他人中,过度表达会加剧呼吸和侵略。PGC1α与不同的信号通路和转录因子的相互作用进一步使其作用多样化,通常以组织特异性的方式。了解这些多方面的功能可以解锁创新的治疗策略。然而,在管理癌细胞的代谢适应性和完善PGC1α靶向方法方面存在挑战。这篇综述旨在整理和介绍当前关于表达模式的知识,监管者,有约束力的合作伙伴,以及PGC1α在多种癌症中的作用。我们检查了PGC1α的组织特异性功能,并阐明了其作为潜在肿瘤抑制因子和致癌合作者的双重性质。在PGC1α抑制肿瘤的癌症中,恢复其水平可以阻止细胞增殖和侵袭,让细胞更容易接受化疗。在相反的癌症中,停止PGC1α的上调可能是有益的,因为它促进氧化磷酸化,允许癌细胞适应压力,并促进更具侵略性的癌症表型。因此,为了有效地瞄准PGC1α,了解其在每种癌症亚型中的细微差别作用是不可或缺的。这可以为肿瘤学领域的重大进展铺平道路。
    PGC1α, a central player in mitochondrial biology, holds a complex role in the metabolic shifts seen in cancer cells. While its dysregulation is common across major cancers, its impact varies. In some cases, downregulation promotes aerobic glycolysis and progression, whereas in others, overexpression escalates respiration and aggression. PGC1α\'s interactions with distinct signaling pathways and transcription factors further diversify its roles, often in a tissue-specific manner. Understanding these multifaceted functions could unlock innovative therapeutic strategies. However, challenges exist in managing the metabolic adaptability of cancer cells and refining PGC1α-targeted approaches. This review aims to collate and present the current knowledge on the expression patterns, regulators, binding partners, and roles of PGC1α in diverse cancers. We examined PGC1α\'s tissue-specific functions and elucidated its dual nature as both a potential tumor suppressor and an oncogenic collaborator. In cancers where PGC1α is tumor-suppressive, reinstating its levels could halt cell proliferation and invasion, and make the cells more receptive to chemotherapy. In cancers where the opposite is true, halting PGC1α\'s upregulation can be beneficial as it promotes oxidative phosphorylation, allows cancer cells to adapt to stress, and promotes a more aggressive cancer phenotype. Thus, to target PGC1α effectively, understanding its nuanced role in each cancer subtype is indispensable. This can pave the way for significant strides in the field of oncology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号