iron metabolism

铁代谢
  • 文章类型: Journal Article
    与代谢功能障碍相关的脂肪肝(MAFLD)相关的复杂病因,包括扰动的铁稳态,它们对疾病进展的贡献性质不明确,导致有效的治疗干预措施数量有限。这里,我们报告代谢功能障碍相关脂肪性肝炎(MASH)患者,MAFLD的病理亚型,表现出过量的肝铁,它与疾病进展有很强的正相关。与临床批准的铁螯合剂相比,FerroTerminator1(FOT1)可有效逆转多种MASH模型的肝损伤,无明显的毒副作用。机械上,我们的多组学分析显示,在各种MASH模型中,FOT1同时抑制肝铁积累和c-Myc-Acsl4触发的铁细胞凋亡.此外,MAFLD队列研究表明,血清铁蛋白水平可能作为MASH中基于FOT1的治疗的预测性生物标志物。这些发现提供了令人信服的证据来支持FOT1作为MAFLD的所有阶段和未来临床试验的有希望的新型治疗选择。
    The complex etiological factors associated with metabolic dysfunction-associated fatty liver disease (MAFLD), including perturbed iron homeostasis, and the unclear nature by which they contribute to disease progression have resulted in a limited number of effective therapeutic interventions. Here, we report that patients with metabolic dysfunction-associated steatohepatitis (MASH), a pathological subtype of MAFLD, exhibit excess hepatic iron and that it has a strong positive correlation with disease progression. FerroTerminator1 (FOT1) effectively reverses liver injury across multiple MASH models without notable toxic side effects compared with clinically approved iron chelators. Mechanistically, our multi-omics analyses reveal that FOT1 concurrently inhibits hepatic iron accumulation and c-Myc-Acsl4-triggered ferroptosis in various MASH models. Furthermore, MAFLD cohort studies suggest that serum ferritin levels might serve as a predictive biomarker for FOT1-based therapy in MASH. These findings provide compelling evidence to support FOT1 as a promising novel therapeutic option for all stages of MAFLD and for future clinical trials.
    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
    炎症性肠病(IBD)是复杂的疾病。铁积聚在IBD患者发炎的组织中,然而,对于炎症的积累机制及其对炎症过程的影响尚不清楚。我们假设炎症改变了铁稳态,影响组织铁的分布,这反过来又延续了炎症。这项研究分析了人体活检,动物模型和细胞系统破译铁稳态在IBD中的作用。我们发现炎症介导的铁分布改变,和铁调节蛋白(IRP)1的铁解偶联激活。为了了解IRP1在这种炎症相关铁模式过程中的作用,建立了一种新的细胞共培养模型,复制了体内观察到的铁模式,并支持一氧化氮参与IRP1的激活和炎症中典型的铁模式。重要的是,从IBD小鼠模型中删除IRP1,铁的分布与肠道炎症。这些发现表明,IRP1在肠粘膜炎症反应的协调中起着核心作用,并且它是IBD治疗干预的可行候选者。
    Inflammatory bowel diseases (IBD) are complex disorders. Iron accumulates in the inflamed tissue of IBD patients, yet neither a mechanism for the accumulation nor its implication on the course of inflammation are known. We hypothesized that the inflammation modifies iron homeostasis, affects tissue iron distribution and that this in turn perpetuates the inflammation. This study analyzed human biopsies, animal models and cellular systems to decipher the role of iron homeostasis in IBD. We found inflammation-mediated modifications of iron distribution, and iron-decoupled activation of the iron regulatory protein (IRP)1. To understand the role of IRP1 in the course of this inflammation-associated iron pattern, a novel cellular co-culture model was established, that replicated the iron-pattern observed in vivo, and supported involvement of nitric oxide in the activation of IRP1 and the typical iron pattern in inflammation. Importantly, deletion of IRP1 from an IBD mouse model completely abolished both, the misdistribution of iron and intestinal inflammation. These findings suggest that IRP1 plays a central role in the coordination of the inflammatory response in the intestinal mucosa and that it is a viable candidate for therapeutic intervention in IBD.
    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
    铁代谢已成为癌症治疗的一个有希望的靶点;然而,癌细胞固有的代谢代偿能力显著限制了铁代谢疗法的有效性。在这里,生物活性硫化镓纳米点(GaSx),具有“重编程”和“干扰”铁代谢途径的双重功能,已成功开发用于肿瘤铁代谢疗法。构造的GaSx纳米点巧妙地利用了硫化氢(H2S)气体,它是响应肿瘤微环境而释放的,重新编程癌细胞中固有的转铁蛋白受体1(TfR1)-铁转运蛋白1(FPN1)铁代谢轴。同时,来自GaSx的镓离子(Ga3+)充当生化“特洛伊木马”,模仿铁的作用并将其从必需的生物分子结合位点置换出来,从而影响癌细胞的命运。通过利用Ga3+介导的铁破坏和H2S促进的铁代谢途径重编程的双重机制,GaSx促进了癌细胞中凋亡-凋亡混合途径的启动,导致肿瘤增殖的显著抑制。重要的是,GaSx诱导的铁代谢失调显著增加了肿瘤细胞对化疗和免疫检查点阻断(ICB)治疗的敏感性.这项研究强调了基于气体的干预措施和金属离子干扰策略对肿瘤代谢治疗的治疗前景。
    Iron metabolism has emerged as a promising target for cancer therapy; however, the innate metabolic compensatory capacity of cancer cells significantly limits the effectiveness of iron metabolism therapy. Herein, bioactive gallium sulfide nanodots (GaSx), with dual functions of \"reprogramming\" and \"interfering\" iron metabolic pathways, were successfully developed for tumor iron metabolism therapy. The constructed GaSx nanodots ingeniously harness hydrogen sulfide (H2S) gas, which is released in response to the tumor microenvironment, to reprogram the inherent transferrin receptor 1 (TfR1)-ferroportin 1 (FPN1) iron metabolism axis in cancer cells. Concurrently, the gallium ions (Ga3+) derived from GaSx act as a biochemical \"Trojan horse\", mimicking the role of iron and displacing it from essential biomolecular binding sites, thereby influencing the fate of cancer cells. By leveraging the dual mechanisms of Ga3+-mediated iron disruption and H2S-facilitated reprogramming of iron metabolic pathways, GaSx prompted the initiation of a paraptosis-apoptosis hybrid pathway in cancer cells, leading to marked suppression of tumor proliferation. Importantly, the dysregulation of iron metabolism induced by GaSx notably increased tumor cell susceptibility to both chemotherapy and immune checkpoint blockade (ICB) therapy. This study underscores the therapeutic promise of gas-based interventions and metal ion interference strategies for the tumor metabolism 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
    生物碱具有显著的生物学和药理学性质,最近受到广泛关注。各种生物碱,包括市售药物如小檗碱,实质上影响铁性凋亡。除了铁死亡的三个主要途径,铁代谢,磷脂代谢,和谷胱甘肽过氧化物酶4调节途径,铁死亡的新机制不断被发现。生物碱可以通过铁凋亡调节各种疾病的进展,并表现出根据剂量和组织类型发挥不同作用的能力,这突显了它们的多功能性。因此,这篇综述全面总结了生物碱在铁凋亡中的主要作用靶点和最新进展,以及生物碱在抑制和促进铁凋亡中的双重作用。
    Alkaloids have remarkable biological and pharmacological properties and have recently garnered extensive attention. Various alkaloids, including commercially available drugs such as berberine, substantially affect ferroptosis. In addition to the three main pathways of ferroptosis, iron metabolism, phospholipid metabolism, and the glutathione peroxidase 4-regulated pathway, novel mechanisms of ferroptosis are continuously being identified. Alkaloids can modulate the progression of various diseases through ferroptosis and exhibit the ability to exert varied effects depending on dosage and tissue type underscores their versatility. Therefore, this review comprehensively summarizes primary targets and the latest advancements of alkaloids in ferroptosis, as well as the dual roles of alkaloids in inhibiting and promoting ferroptosis.
    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
    Ferroptosis,一种以铁依赖性磷脂过氧化为特征的调节性细胞死亡形式,已经成为癌症治疗领域的焦点。与其他细胞死亡模式如凋亡和坏死相比,铁凋亡在分子机制和细胞形态学上表现出许多不同的特征,提供了一个有希望的途径来对抗对传统治疗方式有抵抗力的癌症。鉴于与当前使用外源性铁基无机纳米材料的Fenton调节铁性凋亡疗法相关的严重副作用,劫持内源性铁可以作为一种有效的替代策略,通过靶向细胞铁调节机制来触发铁凋亡。对铁凋亡过程中潜在的铁调节机制的更好理解已经揭示了基于内源性铁凋亡的癌症治疗纳米医学策略的最新发现。在这篇评论文章中,我们对铁代谢的调节网络及其在铁凋亡中的关键作用进行了全面的讨论,并介绍了具有劫持内源性铁以实现铁凋亡能力的纳米颗粒的最新应用。我们设想,该研究中的见解可能会加速内源性基于铁凋亡的纳米药物的开发和翻译,以用于有效的癌症治疗。
    Ferroptosis, a form of regulated cell death characterized by iron-dependent phospholipid peroxidation, has emerged as a focal point in the field of cancer therapy. Compared with other cell death modes such as apoptosis and necrosis, ferroptosis exhibits many distinct characteristics in the molecular mechanisms and cell morphology, offering a promising avenue for combating cancers that are resistant to conventional therapeutic modalities. In light of the serious side effects associated with current Fenton-modulating ferroptosis therapies utilizing exogenous iron-based inorganic nanomaterials, hijacking endogenous iron could serve as an effective alternative strategy to trigger ferroptosis through targeting cellular iron regulatory mechanisms. A better understanding of the underlying iron regulatory mechanism in the process of ferroptosis has shed light on the current findings of endogenous ferroptosis-based nanomedicine strategies for cancer therapy. Here in this review article, we provide a comprehensive discussion on the regulatory network of iron metabolism and its pivotal role in ferroptosis, and present recent updates on the application of nanoparticles endowed with the ability to hijack endogenous iron for ferroptosis. We envision that the insights in the study may expedite the development and translation of endogenous ferroptosis-based nanomedicines for effective cancer 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
    铁代谢在人体的各种生理功能中起着至关重要的作用,因为它对几乎所有生物的生长和发育都至关重要。铁代谢失调-表现为铁缺乏或超负荷-是心血管疾病(CVD)发展的重要危险因素。此外,新出现的证据表明铁性死亡,一种依赖铁的程序性细胞死亡,也可能有助于CVD的发展。了解CVD中铁代谢和铁凋亡的调节机制对于改善疾病管理很重要。通过整合CVD相关铁代谢领域的不同观点和专业知识,本概述提供了有关铁代谢和CVD的见解,以及诊断方法,治疗,并预防与铁失调相关的CVD。
    Iron metabolism plays a crucial role in various physiological functions of the human body, as it is essential for the growth and development of almost all organisms. Dysregulated iron metabolism-manifested either as iron deficiency or overload-is a significant risk factor for the development of cardiovascular disease (CVD). Moreover, emerging evidence suggests that ferroptosis, a form of iron-dependent programed cell death, may also contribute to CVD development. Understanding the regulatory mechanisms of iron metabolism and ferroptosis in CVD is important for improving disease management. By integrating different perspectives and expertise in the field of CVD-related iron metabolism, this overview provides insights into iron metabolism and CVD, along with approaches for diagnosing, treating, and preventing CVD associated with iron dysregulation.
    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
    铁是我们身体的重要微量元素,它的不平衡会导致各种疾病。代谢相关脂肪性肝病(MAFLD)的进展通常伴有铁代谢紊乱。据报道,泽泻提取物(AOE)可缓解MAFLD。然而,关于其特定脂质代谢靶标及其在MAFLD进展过程中对铁代谢的潜在影响的研究仍然有限。建立MAFLD模型,给小鼠喂食标准饮食(CON)或高脂肪饮食(HFD)9周.然后将在HFD上营养的小鼠随机分配到HF组和HFA组,HFA组每天通过管饲法接受AOE,持续13周。补充AOE可显着减少肝脏中过多的脂质积累,并恢复肝脏的铁含量。AOE部分但显著逆转了失调的脂质代谢基因(SCD1,PPARγ,和CD36)和铁代谢基因(TFR1,FPN,和HAMP)由HFD诱导。染色质免疫沉淀测定表明,由HFD诱导的FXR在SCD1和FPN基因启动子上的富集减少被AOE显着逆转。这些发现表明AOE可能通过FXR介导的基因抑制减轻HFD诱导的肝脏脂质和铁代谢紊乱。
    Iron is a vital trace element for our bodies and its imbalance can lead to various diseases. The progression of metabolic-associated fatty liver disease (MAFLD) is often accompanied by disturbances in iron metabolism. Alisma orientale extract (AOE) has been reported to alleviate MAFLD. However, research on its specific lipid metabolism targets and its potential impact on iron metabolism during the progression of MAFLD remains limited. To establish a model of MAFLD, mice were fed either a standard diet (CON) or a high-fat diet (HFD) for 9 weeks. The mice nourished on the HFD were then randomly assigned to the HF group and the HFA group, with the HFA group receiving AOE by gavage on a daily basis for 13 weeks. Supplementation with AOE remarkably reduced overabundant lipid accumulation in the liver and restored the iron content of the liver. AOE partially but significantly reversed dysregulated lipid metabolizing genes (SCD1, PPAR γ, and CD36) and iron metabolism genes (TFR1, FPN, and HAMP) induced by HFD. Chromatin immunoprecipitation assays indicated that the reduced enrichment of FXR on the promoters of SCD1 and FPN genes induced by HFD was significantly reversed by AOE. These findings suggest that AOE may alleviate HFD-induced disturbances in liver lipid and iron metabolism through FXR-mediated gene repression.
    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
    卵巢癌(OC),以其杀伤力和对化疗的抵抗力而闻名,与铁代谢和铁死亡密切相关-铁依赖的细胞死亡过程,有别于自噬和凋亡。新出现的证据表明,铁代谢的失调可能通过诱导氧化还原系统的失衡在OC中起关键作用,这导致了铁性凋亡,提供了一种新的治疗方法。这篇综述探讨了铁代谢的中断,影响氧化还原平衡,影响OC进展,专注于其基本的细胞功能和作为治疗靶标的潜力。它突出了分子的相互作用,包括非编码RNA(ncRNAs)的作用,在铁代谢和铁死亡之间,探索它们与关键免疫细胞如巨噬细胞和T细胞的相互作用,以及肿瘤微环境中的炎症。该综述还讨论了糖酵解相关的铁代谢如何通过活性氧物种影响铁凋亡。瞄准这些途径,特别是通过调节铁代谢和铁凋亡的药物,提出了有希望的治疗前景。该综述强调需要更深入地了解氧化还原调节系统中的铁代谢和铁死亡,以增强OC治疗,并主张继续研究这些机制作为对抗OC的潜在策略。
    Ovarian cancer (OC), known for its lethality and resistance to chemotherapy, is closely associated with iron metabolism and ferroptosis-an iron-dependent cell death process, distinct from both autophagy and apoptosis. Emerging evidence suggests that dysregulation of iron metabolism could play a crucial role in OC by inducing an imbalance in the redox system, which leads to ferroptosis, offering a novel therapeutic approach. This review examines how disruptions in iron metabolism, which affect redox balance, impact OC progression, focusing on its essential cellular functions and potential as a therapeutic target. It highlights the molecular interplay, including the role of non-coding RNAs (ncRNAs), between iron metabolism and ferroptosis, and explores their interactions with key immune cells such as macrophages and T cells, as well as inflammation within the tumor microenvironment. The review also discusses how glycolysis-related iron metabolism influences ferroptosis via reactive oxygen species. Targeting these pathways, especially through agents that modulate iron metabolism and ferroptosis, presents promising therapeutic prospects. The review emphasizes the need for deeper insights into iron metabolism and ferroptosis within the redox-regulated system to enhance OC therapy and advocates for continued research into these mechanisms as potential strategies to combat OC.
    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
    这篇文献综述强调了铁性凋亡在癌症治疗中的创新作用。Ferroptosis是一种故意的细胞死亡,其特征在于脂质过氧化物的产生并且需要铁的存在。Ferroptosis是一种受控的细胞死亡过程,遵守某些规则和规定。系统Xc-的抑制和GPX4的参与是参与铁死亡过程的两个主要探索领域。这篇综述探讨了用于治疗一系列恶性肿瘤中的铁性凋亡的治疗方法。特别关注乳腺癌。注意某些途径,例如谷胱甘肽的FSP1独立调节,胆固醇的参与,和突出蛋白2-MVB/外泌体-铁蛋白途径。Ferroptosis在抵抗肿瘤治疗中起关键作用。
    This literature review emphasizes the innovative role of ferroptosis in cancer treatment. Ferroptosis is a kind of deliberate cell death that is characterized by the generation of lipid peroxides and needs the presence of iron. Ferroptosis is a controlled cell death process that adheres to certain rules and regulations. The inhibition of System Xc- and the involvement of GPX4 are two of the primary areas of exploration that are engaged in the process of ferroptosis. This review explores the treatments that are used to treat ferroptosis in a range of malignancies, with a particular focus on breast carcinoma. Attention is paid to certain pathways, such as the FSP1-independent regulation of glutathione, involvement of cholesterol, and the prominin 2-MVB/exosome-ferritin pathway. Ferroptosis plays a key role in resistance to tumor therapy.
    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
    多柔比星(DOX)是一种用于化疗的抗肿瘤药物,但它的使用往往受到严重和危及生命的心血管毒性副作用的阻碍。近年来,研究集中在铁代谢失调和铁凋亡,铁过载诱导的一种独特类型的细胞死亡,作为推动DOX诱导的心脏毒性(DIC)发展的关键参与者。最近的进展表明,DOX扰乱正常的细胞铁代谢,导致心肌细胞中铁的过度积累和铁凋亡。这篇综述将探讨铁稳态失调和铁凋亡如何驱动DIC的进展。我们还将讨论目前靶向铁代谢和铁凋亡以减轻DIC的方法。此外,我们将讨论这些治疗方案在临床转化方面的局限性和挑战.
    Doxorubicin (DOX) is an anti-tumor agent for chemotherapy, but its use is often hindered by the severe and life-threatening side effect of cardiovascular toxicity. In recent years, studies have focused on dysregulated iron metabolism and ferroptosis, a unique type of cell death induced by iron overload, as key players driving the development of DOX-induced cardiotoxicity (DIC). Recent advances have demonstrated that DOX disturbs normal cellular iron metabolism, resulting in excessive iron accumulation and ferroptosis in cardiomyocytes. This review will explore how dysregulated iron homeostasis and ferroptosis drive the progression of DIC. We will also discuss the current approaches to target iron metabolism and ferroptosis to mitigate DIC. Besides, we will discuss the limitations and challenges for clinical translation for these therapeutic regimens.
    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