Metabolic Reprogramming

代谢重编程
  • 文章类型: Journal Article
    肿瘤相关成纤维细胞(CAFs)是肿瘤微环境(TME)的重要组成部分,它经历了显著的表型变化和代谢重编程,深刻影响肿瘤生长。这篇综述深入研究了CAF可塑性,不同的起源,以及驱动它们持续激活的分子机制。重点放在CAFs和肿瘤细胞之间复杂的双向串扰,促进癌细胞存活,扩散,入侵,和免疫逃避。代谢重编程,癌症的标志,从癌细胞延伸到CAFs,有助于TME内复杂的代谢相互作用。CAF中的“反向Warburg效果”反映了Warburg效果,涉及向癌细胞输出高能底物,支持他们的迅速扩散。像p53,Myc,和K-RAS协调这种代谢适应。了解CAF和肿瘤细胞之间的代谢共生为靶向治疗策略打破这种动态串扰开辟了途径。解开CAF介导的代谢重编程为开发新的抗癌疗法提供了有价值的见解。这项全面的审查巩固了当前的知识,阐明CAFs在TME中的多方面作用,并为未来的治疗提供潜在的靶点。
    Cancer-associated fibroblasts (CAFs) are crucial component of tumor microenvironment (TME) which undergo significant phenotypic changes and metabolic reprogramming, profoundly impacting tumor growth. This review delves into CAF plasticity, diverse origins, and the molecular mechanisms driving their continuous activation. Emphasis is placed on the intricate bidirectional crosstalk between CAFs and tumor cells, promoting cancer cell survival, proliferation, invasion, and immune evasion. Metabolic reprogramming, a cancer hallmark, extends beyond cancer cells to CAFs, contributing to the complex metabolic interplay within the TME. The \'reverse Warburg effect\' in CAFs mirrors the Warburg effect, involving the export of high-energy substrates to fuel cancer cells, supporting their rapid proliferation. Molecular regulations by key players like p53, Myc, and K-RAS orchestrate this metabolic adaptation. Understanding the metabolic symbiosis between CAFs and tumor cells opens avenues for targeted therapeutic strategies to disrupt this dynamic crosstalk. Unraveling CAF-mediated metabolic reprogramming provides valuable insights for developing novel anticancer therapies. This comprehensive review consolidates current knowledge, shedding light on CAFs\' multifaceted roles in the TME and offering potential targets for future therapies.
    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
    肝细胞癌(HCC)是最常见的原发性肝脏恶性肿瘤,由于患病率的增加,其发病率在全球范围内也在增加。代谢重编程已被认为是癌症的标志,并在癌症进展中起作用。葡萄糖,脂质和氨基酸是三种主要成分,它们的代谢改变可以直接影响细胞的能量产生,包括肝癌细胞.营养和能量对于癌细胞的生长和增殖是不可或缺的,因此改变肝癌细胞的代谢可以抑制肝癌的进展。本综述总结了最近对肿瘤调节分子的研究,包括许多非编码RNA,癌基因和肿瘤抑制基因,调节葡萄糖的代谢活动,通过靶向关键酶的脂质和氨基酸,信号通路或两者之间的相互作用。这些调节分子可以调节癌细胞的快速增殖,肿瘤进展和治疗抵抗。据认为,这些肿瘤调节因子可作为肝癌的治疗靶点或有价值的生物标志物。具有减轻HCC耐药性的潜力。此外,代谢抑制剂作为肝癌治疗方法的优缺点,以及可能的解决方案进行了讨论,为开发更有效的HCC治疗策略提供见解。
    Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and its morbidity is increasing worldwide due to increasing prevalence. Metabolic reprogramming has been recognized as a hallmark of cancer and serves a role in cancer progression. Glucose, lipids and amino acids are three major components whose altered metabolism can directly affect the energy production of cells, including liver cancer cells. Nutrients and energy are indispensable for the growth and proliferation of cancer cells, thus altering the metabolism of hepatoma cells can inhibit the progression of HCC. The present review summarizes recent studies on tumour regulatory molecules, including numerous noncoding RNAs, oncogenes and tumour suppressors, which regulate the metabolic activities of glucose, lipids and amino acids by targeting key enzymes, signalling pathways or interactions between the two. These regulatory molecules can regulate the rapid proliferation of cancer cells, tumour progression and treatment resistance. It is thought that these tumour regulatory factors may serve as therapeutic targets or valuable biomarkers for HCC, with the potential to mitigate HCC drug resistance. Furthermore, the advantages and disadvantages of metabolic inhibitors as a treatment approach for HCC, as well as possible solutions are discussed, providing insights for developing more effective treatment strategies for HCC.
    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
    乳酸以前被认为是代谢的副产品。然而,对癌症发展的复杂性的广泛研究揭示了它对肿瘤生长的重要贡献,迁移,和入侵。在组蛋白和非组蛋白蛋白中广泛观察到涉及乳酸的翻译后修饰,这些修饰通过共价连接乳酰和蛋白质中的赖氨酸残基在调节基因表达中起着至关重要的作用。这一发现大大增强了我们对乳酸参与疾病发病机制的理解。在这篇文章中,我们对乳酸与肿瘤免疫之间的复杂关系进行了全面的综述,在恶性肿瘤中发生乳酸化,以及在肿瘤免疫治疗中靶向乳酸-乳酸化的开发。此外,我们讨论了未来的研究方向,旨在提供新颖的见解,可以为调查提供信息,诊断,以及相关疾病的治疗。
    Lactic acid was formerly regarded as a byproduct of metabolism. However, extensive investigations into the intricacies of cancer development have revealed its significant contributions to tumor growth, migration, and invasion. Post-translational modifications involving lactate have been widely observed in histone and non-histone proteins, and these modifications play a crucial role in regulating gene expression by covalently attaching lactoyl groups to lysine residues in proteins. This discovery has greatly enhanced our comprehension of lactic acid\'s involvement in disease pathogenesis. In this article, we provide a comprehensive review of the intricate relationship between lactate and tumor immunity, the occurrence of lactylation in malignant tumors, and the exploitation of targeted lactate-lactylation in tumor immunotherapy. Additionally, we discuss future research directions, aiming to offer novel insights that could inform the investigation, diagnosis, and treatment of related 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
    妊娠期糖尿病是一种常见的代谢性疾病,可影响妊娠和分娩的正常过程。导致母亲和孩子的不良后果。其发病机制复杂,涉及多种因素,如胰岛素抵抗和β细胞功能障碍。代谢重编程,涉及线粒体氧化磷酸化和糖酵解,对于维持人体代谢平衡至关重要,并且与妊娠期糖尿病的发病机制和进展有关。然而,关于代谢重编程与妊娠期糖尿病之间的联系和代谢途径的研究是有限的。因此,我们综述了代谢重编程与妊娠期糖尿病之间的关系,以期为妊娠期孕产妇健康提供新的治疗策略,降低妊娠期糖尿病的风险。
    Gestational diabetes mellitus is a prevalent metabolic disease that can impact the normal course of pregnancy and delivery, leading to adverse outcomes for both mother and child. Its pathogenesis is complex and involves various factors, such as insulin resistance and β-cell dysfunction. Metabolic reprogramming, which involves mitochondrial oxidative phosphorylation and glycolysis, is crucial for maintaining human metabolic balance and is involved in the pathogenesis and progression of gestational diabetes mellitus. However, research on the link and metabolic pathways between metabolic reprogramming and gestational diabetes mellitus is limited. Therefore, we reviewed the relationship between metabolic reprogramming and gestational diabetes mellitus to provide new therapeutic strategies for maternal health during pregnancy and reduce the risk of developing gestational diabetes mellitus.
    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
    随着近年来对m6A相关机制研究的进展,m6A阅读器中的YTHDF蛋白家族引起了极大的关注。其中,YTHDF1作为关键成员,在蛋白质翻译中起着至关重要的作用,肿瘤增殖,各种肿瘤细胞的代谢重编程,和免疫逃避。此外,YTHDF1还通过多种信号通路对肿瘤发挥调节作用,和许多研究已经证实了它的能力,以协助重编程的肿瘤细胞相关的代谢过程。近年来,对YTHDF1的研究重点已从其m6A识别和修饰功能转移到其调节肿瘤进展的分子机制。特别是通过探索与YTHDF1上游和下游相互作用的调节因素。在这次审查中,我们阐明了YTHDF1在各种肿瘤细胞中的最新信号通路机制,特别强调其在肿瘤细胞代谢重编程中的独特特征。此外,我们总结了YTHDF1在肿瘤细胞中的最新病理和生理过程,并分析利用YTHDF1的潜在治疗方法。我们相信YTHDF1代表了未来肿瘤治疗的一个非常有希望的靶标和新的肿瘤生物标志物。
    With the advancement of research on m6A-related mechanisms in recent years, the YTHDF protein family within m6A readers has garnered significant attention. Among them, YTHDF1 serves as a pivotal member, playing a crucial role in protein translation, tumor proliferation, metabolic reprogramming of various tumor cells, and immune evasion. In addition, YTHDF1 also exerts regulatory effects on tumors through multiple signaling pathways, and numerous studies have confirmed its ability to assist in the reprogramming of the tumor cell-related metabolic processes. The focus of research on YTHDF1 has shifted in recent years from its m6A-recognition and -modification function to the molecular mechanisms by which it regulates tumor progression, particularly by exploring the regulatory factors that interact with YTHDF1 upstream and downstream. In this review, we elucidate the latest signaling pathway mechanisms of YTHDF1 in various tumor cells, with a special emphasis on its distinctive characteristics in tumor cell metabolic reprogramming. Furthermore, we summarize the latest pathological and physiological processes involving YTHDF1 in tumor cells, and analyze potential therapeutic approaches that utilize YTHDF1. We believe that YTHDF1 represents a highly promising target for future tumor treatments and a novel tumor biomarker.
    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
    肝细胞癌(HCC)对人类健康构成了沉重的负担,具有很高的发病率和死亡率。系统治疗对于晚期和中期HCC至关重要,但面临治疗抗性的重大挑战,削弱药物的有效性。代谢重编程作为治疗抗性的关键促成因素而受到关注。细胞改变新陈代谢来满足能量需求,适应成长需要,或抵抗环境压力。了解关键的酶表达模式和代谢途径相互作用对于理解HCC的发生至关重要。发展,和治疗阻力。探索代谢酶重编程和途径对于确定HCC治疗的突破口至关重要。用抑制剂靶向代谢酶是解决这些问题的关键。抑制剂,结合全身治疗药物,可以减轻抵抗力,延长晚期肝癌的总生存期,并为中期肝癌患者提供根治性切除的机会。代谢研究方法的进展,从基因组学到代谢组学,从细胞到类器官,帮助建立HCC代谢重编程网络。生物材料和纳米技术的最新进展影响药物靶向和有效性,为系统治疗性耐药提供新的解决方案。本文综述了代谢酶的变化,途径相互作用,酶抑制剂,研究方法,和靶向代谢重编程的药物递送,为肝癌的代谢治疗方法提供有价值的参考。
    Hepatocellular carcinoma (HCC) poses a heavy burden on human health with high morbidity and mortality rates. Systematic therapy is crucial for advanced and mid-term HCC, but faces a significant challenge from therapeutic resistance, weakening drug effectiveness. Metabolic reprogramming has gained attention as a key contributor to therapeutic resistance. Cells change their metabolism to meet energy demands, adapt to growth needs, or resist environmental pressures. Understanding key enzyme expression patterns and metabolic pathway interactions is vital to comprehend HCC occurrence, development, and treatment resistance. Exploring metabolic enzyme reprogramming and pathways is essential to identify breakthrough points for HCC treatment. Targeting metabolic enzymes with inhibitors is key to addressing these points. Inhibitors, combined with systemic therapeutic drugs, can alleviate resistance, prolong overall survival for advanced HCC, and offer mid-term HCC patients a chance for radical resection. Advances in metabolic research methods, from genomics to metabolomics and cells to organoids, help build the HCC metabolic reprogramming network. Recent progress in biomaterials and nanotechnology impacts drug targeting and effectiveness, providing new solutions for systemic therapeutic drug resistance. This review focuses on metabolic enzyme changes, pathway interactions, enzyme inhibitors, research methods, and drug delivery targeting metabolic reprogramming, offering valuable references for metabolic approaches to HCC treatment.
    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
    单细胞转录组学的联合分析,蛋白质组学,脂质组学,代谢组学和空间代谢组学正在不断改变我们对肿瘤细胞代谢重编程机制的理解。由于头颈部肿瘤是世界上第六大最常见的肿瘤,对其发生的代谢机制的研究,发展和预后仍然不发达。在过去的十年里,该领域经历了巨大的技术革命和长足的发展,使人类肿瘤代谢研究取得了重大突破。在这次审查中,对传统代谢组学和空间代谢组学进行了全面比较,并对空间代谢组学联合多组学在肿瘤代谢重编程研究中的应用进展和面临的挑战进行了综述。此外,我们还强调了空间代谢组学在头颈部肿瘤代谢机制研究中的进展,并对其应用前景进行了展望。
    The joint analysis of single-cell transcriptomics, proteomics, lipidomics, metabolomics and spatial metabolomics is continually transforming our understanding of the mechanisms of metabolic reprogramming in tumor cells. Since head and neck tumor is the sixth most common tumor in the world, the study of the metabolic mechanism of its occurrence, development and prognosis is still undeveloped. In the past decade, this field has witnessed tremendous technological revolutions and considerable development that enables major breakthroughs to be made in the study of human tumor metabolism. In this review, a comprehensive comparison of traditional metabolomics and spatial metabolomics has been concluded, and the recent progress and challenges of the application of spatial metabolomics combined multi-omics in the research of metabolic reprogramming in tumors are reviewed. Furthermore, we also highlight the advances of spatial metabolomics in the study of metabolic mechanisms of head and neck tumors, and provide an outlook of its application prospects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    膀胱癌(BLCA)是最常见的尿路上皮癌。代谢重编程在肿瘤中的作用逐渐受到关注和研究。最近的研究表明,非编码RNA(ncRNA)与BLCA代谢重编程密切相关。ncRNAs能够直接调节代谢酶的表达和功能,或通过许多重要途径间接调节它们以调节BLCA细胞的代谢。BLCA的发展机制还没有,据作者所知,已经研究和鉴定ncRNAs在BLCA代谢重编程中的作用可能有助于开发BLCA治疗。在本次审查中,提供了参与BLCA代谢重编程的ncRNAs的总结.葡萄糖中ncRNAs的调节,还详细介绍了脂质和氨基酸代谢。此外,我们讨论了BLCA代谢重编程中ncRNAs调控的分子机制,以及通过调控特定ncRNAs的表达来追踪癌细胞代谢的潜在治疗策略.
    Bladder cancer (BLCA) is the most common type of urothelial cancer. The role of metabolic reprogramming in tumors is gradually gaining more attention and being investigated. Recent studies have shown that noncoding RNAs (ncRNAs) are strongly related to BLCA metabolic reprogramming. ncRNAs are able to directly regulate the expression and function of metabolic enzymes, or indirectly regulate them through a number of important pathways to regulate metabolism in BLCA cells. The mechanism of the development of BLCA has not yet, to the best of the authors\' knowledge, been studied and identifying how ncRNAs act in metabolic reprogramming in BLCA may assist with developing BLCA treatments. In the present review, a summary of the ncRNAs participating in the metabolic reprogramming of BLCA was provided. The regulation of ncRNAs in glucose, lipid and amino acid metabolism was also detailed. Furthermore, the molecular mechanisms underlying the regulation of ncRNAs in the metabolic reprogramming of BLCA and potential treatment strategies that track cancer cell metabolism by regulating the expression of particular ncRNAs were discussed.
    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
    结直肠癌(CRC)是一个主要的健康问题,多因素病理生理学代表着强烈的治疗挑战。众所周知,时空控制的信号通路的失调及其代谢重编程效应在CRC的发生和发展中起关键作用。因此,线粒体在CRC启动中的作用最近获得了很多关注,因为它被认为是调节CRC中生物能学的动力。此外,microRNAs(miRNAs)与线粒体功能障碍之间的串扰已成为破译CRC分子机制的新热情。本文综述了参与代谢重编程的不同信号通路与其治疗靶点之间的关系。线粒体DNA含量的改变,线粒体生物发生,和线粒体自噬,和线粒体基因多态性的作用以及调节线粒体蛋白的miRNA在CRC起始,programming,转移,以及对各种疗法的抵制。
    Colorectal cancer (CRC) is a major health concern with multifactorial pathophysiology representing intense therapeutic challenges. It is well known that deregulation of spatiotemporally-controlled signaling pathways and their metabolic reprogramming effects play a pivotal role in the development and progression of CRC. As such, the mitochondrial role in CRC initiation gained a lot of attention recently, as it is considered the powerhouse that regulates the bioenergetics in CRC. In addition, the crosstalk between microRNAs (miRNAs) and mitochondrial dysfunction has become a newfangled passion for deciphering CRC molecular mechanisms. This review sheds light on the relationship between different signaling pathways involved in metabolic reprogramming and their therapeutic targets, alterations in mitochondrial DNA content, mitochondrial biogenesis, and mitophagy, and the role of polymorphisms in mitochondrial genes as well as miRNAs regulating mitochondrial proteins in CRC initiation, progression, metastasis, and resistance to various therapies.
    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
    肝纤维化可能是治疗肝癌的最后希望,肝微环境的重塑已成为促进肝纤维化消融的策略。近年来,特别是随着纳米医学的快速发展,肝微环境治疗在肝癌和纤维化的研究中得到了广泛的研究。在这次全面审查中,我们总结了基于纳米疗法的肝微环境重塑的最新进展。首先,我们讨论了由肝窦内皮细胞(LSEC)毛细血管化和巨噬细胞极化引起的调节性免疫抑制的新策略。此外,代谢重编程和细胞外基质(ECM)沉积是由肝星状细胞(HSC)的激活引起的。此外,ROS的最新进展,缺氧,还总结了由于ECM沉积导致的肝纤维化微环境中血管重塑受损。最后,这篇综述讨论了基于相关信号的新兴纳米治疗方法。我们已经提出了新的策略,如工程纳米治疗靶向抗原呈递细胞(APC)或直接靶向T细胞在肝纤维化免疫疗法中用于预防肝纤维化。总之,这篇全面的综述说明了药物靶向和纳米医学的机遇,以及当前需要解决的挑战。
    Liver fibrosis could be the last hope for treating liver cancer and remodeling of the hepatic microenvironment has emerged as a strategy to promote the ablation of liver fibrosis. In recent years, especially with the rapid development of nanomedicine, hepatic microenvironment therapy has been widely researched in studies concerning liver cancer and fibrosis. In this comprehensive review, we summarized recent advances in nano therapy-based remodeling of the hepatic microenvironment. Firstly, we discussed novel strategies for regulatory immune suppression caused by capillarization of liver sinusoidal endothelial cells (LSECs) and macrophage polarization. Furthermore, metabolic reprogramming and extracellular matrix (ECM) deposition are caused by the activation of hepatic stellate cells (HSCs). In addition, recent advances in ROS, hypoxia, and impaired vascular remodeling in the hepatic fibrotic microenvironment due to ECM deposition have also been summarized. Finally, emerging nanotherapeutic approaches based on correlated signals were discussed in this review. We have proposed novel strategies such as engineered nanotherapeutics targeting antigen-presenting cells (APCs) or direct targeting T cells in liver fibrotic immunotherapy to be used in preventing liver fibrosis. In summary, this comprehensive review illustrated the opportunities in drug targeting and nanomedicine, and the current challenges to be addressed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号