cancer metabolism

癌症代谢
  • 文章类型: Journal Article
    在全球范围内,肺癌是癌症相关死亡率的主要原因。尽管在治疗干预方面取得了进展,包括外科手术,辐射,化疗,靶向治疗和免疫疗法,总体预后仍然不利。氧化还原平衡失衡和氧化还原信号中断,肿瘤的共同特征,在恶性进展和治疗抵抗中起关键作用。癌细胞,通常以遗传引起的持续高水平的活性氧(ROS)为特征,新陈代谢,和微环境的改变,通过增强它们的抗氧化能力来抵消这一点。半胱氨酸的可用性成为化学抗性的关键因素,塑造非小细胞肺癌(NSCLC)细胞的生存动态。公开了硒-chrysin(SeChry)作为半胱氨酸细胞内可用性的调节剂。这项研究全面表征了SeChry的代谢,并研究了其在NSCLC中的细胞毒性作用。SeChry处理诱导显著的代谢变化,特别是在硒化合物代谢中,影响关键途径,如糖酵解,糖异生,三羧酸(TCA)循环,和氨基酸代谢。此外,SeChry影响关键代谢物的水平,如乙酸盐,乳酸,葡萄糖,和氨基酸,导致氧化还原稳态和细胞生物合成的破坏。SeChry与其他治疗方法的结合,如糖酵解抑制和化疗,导致更大的疗效。此外,通过利用非小细胞肺癌消耗乳酸的能力,研究了乳酸共轭树枝状聚合物纳米颗粒用于SeChry递送的用途,对表达单羧酸转运蛋白的癌细胞显示特异性。
    Lung cancer ranks as the predominant cause of cancer-related mortalities on a global scale. Despite progress in therapeutic interventions, encompassing surgical procedures, radiation, chemotherapy, targeted therapies and immunotherapy, the overall prognosis remains unfavorable. Imbalances in redox equilibrium and disrupted redox signaling, common traits in tumors, play crucial roles in malignant progression and treatment resistance. Cancer cells, often characterized by persistent high levels of reactive oxygen species (ROS) resulting from genetic, metabolic, and microenvironmental alterations, counterbalance this by enhancing their antioxidant capacity. Cysteine availability emerges as a critical factor in chemoresistance, shaping the survival dynamics of non‑small cell lung cancer (NSCLC) cells. Selenium-chrysin (SeChry) was disclosed as a modulator of cysteine intracellular availability. This study comprehensively characterizes the metabolism of SeChry and investigates its cytotoxic effects in NSCLC. SeChry treatment induces notable metabolic shifts, particularly in selenocompound metabolism, impacting crucial pathways such as glycolysis, gluconeogenesis, the tricarboxylic acid (TCA) cycle, and amino acid metabolism. Additionally, SeChry affects the levels of key metabolites such as acetate, lactate, glucose, and amino acids, contributing to disruptions in redox homeostasis and cellular biosynthesis. The combination of SeChry with other treatments, such as glycolysis inhibition and chemotherapy, results in greater efficacy. Furthermore, by exploiting NSCLC\'s capacity to consume lactate, the use of lactic acid-conjugated dendrimer nanoparticles for SeChry delivery is investigated, showing specificity to cancer cells expressing monocarboxylate transporters.
    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
    癌症代谢现在是治疗干预的关键领域,靶向独特的代谢重编程对于肿瘤生长和存活至关重要。本文回顾了通过糖酵解和谷氨酰胺酶抑制剂解决代谢脆弱性的治疗潜力,破坏癌细胞的新陈代谢.讨论了肿瘤异质性和适应性抗性等挑战,采用包括个性化医疗和预测生物标志物在内的策略来提高治疗效果。此外,将饮食和生活方式的改变与代谢靶向相结合,强调了改善治疗结果的整体方法.本文还研究了将这些策略纳入标准护理的好处,强调更有可能量身定制,更安全的治疗方法总之,利用代谢漏洞预示着肿瘤学的新时代,将代谢靶向定位在个性化癌症治疗和转变患者护理的最前沿。
    Cancer metabolism is now a key area for therapeutic intervention, targeting unique metabolic reprogramming crucial for tumor growth and survival. This article reviews the therapeutic potential of addressing metabolic vulnerabilities through glycolysis and glutaminase inhibitors, which disrupt cancer cell metabolism. Challenges such as tumor heterogeneity and adaptive resistance are discussed, with strategies including personalized medicine and predictive biomarkers to enhance treatment efficacy. Additionally, integrating diet and lifestyle changes with metabolic targeting underscores a holistic approach to improving therapy outcomes. The article also examines the benefits of incorporating these strategies into standard care, highlighting the potential for more tailored, safer treatments. In conclusion, exploiting metabolic vulnerabilities promises a new era in oncology, positioning metabolic targeting at the forefront of personalized cancer therapy and transforming patient care.
    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
    癌细胞经历代谢重编程以在低氧条件下存活并满足癌症微环境的升高的能量需求。这种代谢改变是由缺氧诱导因子1(HIF-1),调节癌细胞内的各种过程。缺氧诱导的复杂代谢修饰强调了HIF-1诱导的代谢重编程在促进癌症进展的各个方面的重要性。HIF-1信号和细胞代谢过程之间的复杂的相互作用响应缺氧在这项研究中进行了检查,专注于碳水化合物的代谢,核苷酸,脂质,和氨基酸。理解细胞代谢中HIF-1控制的各种调节机制揭示了癌症生长的复杂生物学,并为开发靶向治疗提供了有用的见解。
    Cancer cells undergo metabolic reprogramming to survive in hypoxic conditions and meet the elevated energy demands of the cancer microenvironment. This metabolic alteration is orchestrated by hypoxia-inducible factor 1 (HIF-1), regulating various processes within cancer cells. The intricate metabolic modifications induced by hypoxia underscore the significance of HIF-1-induced metabolic reprogramming in promoting each aspect of cancer progression. The complex interactions between HIF-1 signalling and cellular metabolic processes in response to hypoxia are examined in this study, focusing on the metabolism of carbohydrates, nucleotides, lipids, and amino acids. Comprehending the various regulatory mechanisms controlled by HIF-1 in cellular metabolism sheds light on the intricate biology of cancer growth and offers useful insights for developing targeted treatments.
    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
    铁是生命的基本要素,因为它能够参与各种氧化还原反应。然而,铁依赖性氧化还原循环的失调也会产生氧化应激,有助于细胞生长,扩散,和衰老背后的死亡途径,癌症,神经变性,和代谢性疾病。选择性监测松散结合的Fe(II)离子的荧光探针,被称为不稳定的铁池,是研究这种金属营养素的潜在强大工具;然而,这些生物可利用的金属库的动态时空性质和有效的荧光猝灭能力对其检测提出了挑战。这里,我们报告了一种串联的基于活动的传感和标记策略,该策略通过增强细胞滞留,能够对活细胞中不稳定的铁池进行成像.铁绿-1氟甲基(IG1-FM)使用内过氧化物触发剂与Fe(II)选择性反应,释放醌甲基化染料,随后附着于近端生物亲核试剂,在升高的不稳定铁的部位提供永久性荧光染色。IG1-FM成像显示,主要的铁储存蛋白铁蛋白通过铁吞噬的降解扩大了不稳定的铁库,而核因子-红系2相关因子2(NRF2)抗氧化反应元件(AREs)的激活会耗尽它。我们进一步表明,肺癌细胞具有增强的NRF2激活,从而降低了基础不稳定铁,当用铁螯合剂处理时具有降低的活力。通过将不稳定的铁池和NRF2-ARE活动与癌症中依赖金属的药物脆弱性联系起来,这项工作为更广泛地研究过渡金属和抗氧化剂信号通路在健康和疾病中的作用提供了起点.
    Iron is an essential element for life owing to its ability to participate in a diverse array of oxidation-reduction reactions. However, misregulation of iron-dependent redox cycling can also produce oxidative stress, contributing to cell growth, proliferation, and death pathways underlying aging, cancer, neurodegeneration, and metabolic diseases. Fluorescent probes that selectively monitor loosely bound Fe(II) ions, termed the labile iron pool, are potentially powerful tools for studies of this metal nutrient; however, the dynamic spatiotemporal nature and potent fluorescence quenching capacity of these bioavailable metal stores pose challenges for their detection. Here, we report a tandem activity-based sensing and labeling strategy that enables imaging of labile iron pools in live cells through enhancement in cellular retention. Iron green-1 fluoromethyl (IG1-FM) reacts selectively with Fe(II) using an endoperoxide trigger to release a quinone methide dye for subsequent attachment to proximal biological nucleophiles, providing a permanent fluorescent stain at sites of elevated labile iron. IG1-FM imaging reveals that degradation of the major iron storage protein ferritin through ferritinophagy expands the labile iron pool, while activation of nuclear factor-erythroid 2-related factor 2 (NRF2) antioxidant response elements (AREs) depletes it. We further show that lung cancer cells with heightened NRF2 activation, and thus lower basal labile iron, have reduced viability when treated with an iron chelator. By connecting labile iron pools and NRF2-ARE activity to a druggable metal-dependent vulnerability in cancer, this work provides a starting point for broader investigations into the roles of transition metal and antioxidant signaling pathways in health and disease.
    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
    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
    代谢重编程是癌症的标志,推动靶向癌症代谢疗法的发展。稳定同位素示踪已成为在体外和体内监测癌症代谢的广泛采用的工具。仪器仪表的进步和新型示踪剂的开发,代谢物数据库,和数据分析工具扩大了癌症代谢研究的范围。在这次审查中,我们探索代谢分析的最新进展,从稳定同位素标记代谢组学的实验设计到复杂的数据分析技术。我们强调在癌症研究中的成功应用,特别关注利用稳定同位素示踪来表征疾病进展的正在进行的临床试验,治疗反应,和抗癌疗法耐药的潜在机制。此外,我们概述了关键挑战,并讨论了解决这些挑战的潜在策略,旨在增强我们对癌症代谢的生化基础的理解。
    Metabolic reprogramming is a hallmark of cancer, driving the development of therapies targeting cancer metabolism. Stable isotope tracing has emerged as a widely adopted tool for monitoring cancer metabolism both in vitro and in vivo. Advances in instrumentation and the development of new tracers, metabolite databases, and data analysis tools have expanded the scope of cancer metabolism studies across these scales. In this review, we explore the latest advancements in metabolic analysis, spanning from experimental design in stable isotope-labeling metabolomics to sophisticated data analysis techniques. We highlight successful applications in cancer research, particularly focusing on ongoing clinical trials utilizing stable isotope tracing to characterize disease progression, treatment responses, and potential mechanisms of resistance to anticancer therapies. Furthermore, we outline key challenges and discuss potential strategies to address them, aiming to enhance our understanding of the biochemical basis of cancer metabolism.
    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
    与正常细胞相反,癌细胞主要利用糖酵解在有氧条件下产生ATP,促进增殖和转移。靶向糖酵解对于癌症治疗是有效的。Prodigiosin(PDG)是一种具有多种生物活性的天然化合物,包括抗癌作用。然而,PDG的精确作用机制和分子靶标,已经证明了在调节癌细胞葡萄糖代谢方面的功效,仍然难以捉摸。这里,我们旨在研究PDG的抗癌活性和在癌症代谢中的作用机制。PDG通过抑制细胞内ATP产生速率和水平来调节癌症代谢。它抑制糖酵解和线粒体氧化磷酸化,阻碍ATP的产生依赖于糖酵解和线粒体呼吸。此外,它通过直接与葡萄糖转运蛋白1相互作用而抑制细胞葡萄糖摄取,而不影响其在HCT116细胞中的mRNA或蛋白质水平。我们提供了通过癌症代谢调节介导的PDG的抗癌作用的见解,表明它对癌症的治疗潜力。
    In contrast to normal cells, cancer cells predominantly utilise glycolysis for ATP generation under aerobic conditions, facilitating proliferation and metastasis. Targeting glycolysis is effective for cancer treatment. Prodigiosin (PDG) is a natural compound with various bioactivities, including anticancer effects. However, the precise action mechanisms and molecular targets of PDG, which has demonstrated efficacy in regulating glucose metabolism in cancer cells, remain elusive. Here, we aimed to investigate the anti-cancer activity of PDG and mechanism in cancer metabolism. PDG regulated cancer metabolism by suppressing intracellular ATP production rate and levels. It inhibited glycolysis and mitochondrial oxidative phosphorylation, impeding ATP production dependent on both glycolysis and mitochondrial respiration. Moreover, it inhibited cellular glucose uptake by directly interacting with glucose transporter 1 without affecting its mRNA or protein levels in HCT116 cells. We provide insights into the anti-cancer effects of PDG mediated via cancer metabolism regulation, suggesting its therapeutic potential for 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
    PI3K/Akt途径的异常激活通常发生在癌症中,并且与恶性进展的多个方面相关。特别是,最近的证据表明,PI3K/Akt信号在促进所谓的有氧糖酵解或Warburg效应中起着基本作用,通过磷酸化不同的营养转运蛋白和代谢酶,例如GLUT1,HK2,PFKB3/4和PKM2,并通过调节各种分子网络和蛋白质,包括mTORC1,GSK3,FOXO转录因子,MYC和HIF-1α。这导致癌症代谢的深刻重新编程,也影响磷酸戊糖途径,线粒体氧化磷酸化,从头脂质合成和氧化还原稳态,从而满足肿瘤细胞的分解代谢和合成代谢需求。本综述讨论了PI3K/Akt级联与其代谢靶标之间的相互作用,关注它们可能的治疗意义。
    Aberrant activation of the PI3K/Akt pathway commonly occurs in cancers and correlates with multiple aspects of malignant progression. In particular, recent evidence suggests that the PI3K/Akt signaling plays a fundamental role in promoting the so-called aerobic glycolysis or Warburg effect, by phosphorylating different nutrient transporters and metabolic enzymes, such as GLUT1, HK2, PFKB3/4 and PKM2, and by regulating various molecular networks and proteins, including mTORC1, GSK3, FOXO transcription factors, MYC and HIF-1α. This leads to a profound reprogramming of cancer metabolism, also impacting on pentose phosphate pathway, mitochondrial oxidative phosphorylation, de novo lipid synthesis and redox homeostasis and thereby allowing the fulfillment of both the catabolic and anabolic demands of tumor cells. The present review discusses the interactions between the PI3K/Akt cascade and its metabolic targets, focusing on their possible therapeutic implications.
    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