首页 > 最新文献

Biomedicine & Pharmacotherapy最新文献

英文 中文
Morusin, a novel inhibitor of ACLY, induces mitochondrial apoptosis in hepatocellular carcinoma cells through ROS-mediated mitophagy 新型 ACLY 抑制剂 Morusin 通过 ROS 介导的有丝分裂诱导肝癌细胞线粒体凋亡
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-27 DOI: 10.1016/j.biopha.2024.117510

Objective

Morusin (Mor), a prenylated flavonoid isolated from the root bark of Morus alba L., exhibits potent anti-tumour effects; however, the molecular target of Mor is still not entirely clear. This study aimed to elucidate the mechanism of Mor against hepatocellular carcinoma (HCC) and identify potential molecular targets.

Methods

Mitochondrial function was assessed by measuring the mitochondrial membrane potential, mitochondrial ultrastructure, oxygen consumption, and ATP levels. Mor-induced mitophagy was confirmed using western blotting, immunofluorescence, and fluorescent probes. Transcriptomics, flow cytometry, western blotting, qRT-PCR and biochemical assays were used to reveal the molecular mechanisms and targets of Mor against HCC. We further validated the interaction between Mor and the target proteins using molecular docking and biolayer interferometry (BLI). The inhibitory effect of Mor in vivo was evaluated using a Hep3B murine xenograft model.

Results

Mor significantly reduced the ATP citrate lyase (ACLY) expression and inhibited ACLY activity in HCC cells. BLI analysis demonstrated a direct interaction between Mor and the ACLY active domain. Mor-induced ACLY inhibition led to ROS accumulation in HCC cells, which caused mitochondrial damage, triggered PINK1/Parkin-mediated mitophagy, and ultimately induced mitochondrial apoptosis. We further verified that ROS is crucial in the apoptotic action of Mor through experiments regarding an ROS scavenger. Mor also significantly inhibited tumour xenograft growth in vivo. In addition, analysis of human liver cancer clinical samples revealed elevated ACLY levels positively correlated with histologic grade.

Conclusion

Collectively, our findings highlight Mor as a potent bioactive inhibitor of ACLY and a promising candidate for HCC therapy.
目的Morusin(Mor)是从桑树根皮中分离出来的一种前烯化黄酮类化合物,具有很强的抗肿瘤作用;然而,Mor的分子靶点仍不完全清楚。本研究旨在阐明 Mor 抗肝细胞癌(HCC)的机制,并确定潜在的分子靶点。方法通过测量线粒体膜电位、线粒体超微结构、耗氧量和 ATP 水平来评估线粒体功能。使用 Western 印迹、免疫荧光和荧光探针证实了 Mor 诱导的有丝分裂。我们利用转录组学、流式细胞术、Western 印迹、qRT-PCR 和生化分析揭示了 Mor 抗 HCC 的分子机制和靶点。我们还利用分子对接和生物层干涉仪(BLI)进一步验证了Mor与靶蛋白之间的相互作用。我们使用 Hep3B 小鼠异种移植模型评估了 Mor 在体内的抑制作用。BLI 分析表明 Mor 与 ACLY 活性结构域之间存在直接相互作用。Mor诱导的ACLY抑制导致ROS在HCC细胞中积累,造成线粒体损伤,引发PINK1/Parkin介导的有丝分裂,最终诱导线粒体凋亡。我们通过ROS清除剂的实验进一步验证了ROS在Mor的凋亡作用中的关键作用。Mor 还能明显抑制体内肿瘤异种移植的生长。此外,对人类肝癌临床样本的分析表明,ACLY 水平的升高与组织学分级呈正相关。
{"title":"Morusin, a novel inhibitor of ACLY, induces mitochondrial apoptosis in hepatocellular carcinoma cells through ROS-mediated mitophagy","authors":"","doi":"10.1016/j.biopha.2024.117510","DOIUrl":"10.1016/j.biopha.2024.117510","url":null,"abstract":"<div><h3>Objective</h3><div>Morusin (Mor), a prenylated flavonoid isolated from the root bark of <em>Morus alba L</em>., exhibits potent anti-tumour effects; however, the molecular target of Mor is still not entirely clear. This study aimed to elucidate the mechanism of Mor against hepatocellular carcinoma (HCC) and identify potential molecular targets.</div></div><div><h3>Methods</h3><div>Mitochondrial function was assessed by measuring the mitochondrial membrane potential, mitochondrial ultrastructure, oxygen consumption, and ATP levels. Mor-induced mitophagy was confirmed using western blotting, immunofluorescence, and fluorescent probes. Transcriptomics, flow cytometry, western blotting, qRT-PCR and biochemical assays were used to reveal the molecular mechanisms and targets of Mor against HCC. We further validated the interaction between Mor and the target proteins using molecular docking and biolayer interferometry (BLI). The inhibitory effect of Mor <em>in vivo</em> was evaluated using a Hep3B murine xenograft model.</div></div><div><h3>Results</h3><div>Mor significantly reduced the ATP citrate lyase (ACLY) expression and inhibited ACLY activity in HCC cells. BLI analysis demonstrated a direct interaction between Mor and the ACLY active domain. Mor-induced ACLY inhibition led to ROS accumulation in HCC cells, which caused mitochondrial damage, triggered PINK1/Parkin-mediated mitophagy, and ultimately induced mitochondrial apoptosis. We further verified that ROS is crucial in the apoptotic action of Mor through experiments regarding an ROS scavenger. Mor also significantly inhibited tumour xenograft growth <em>in vivo</em>. In addition, analysis of human liver cancer clinical samples revealed elevated ACLY levels positively correlated with histologic grade.</div></div><div><h3>Conclusion</h3><div>Collectively, our findings highlight Mor as a potent bioactive inhibitor of ACLY and a promising candidate for HCC therapy.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142326359","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nuclear factor erythroid 2-related factor-mediated signaling alleviates ferroptosis during cerebral ischemia-reperfusion injury 核因子红细胞 2 相关因子介导的信号传导缓解了脑缺血再灌注损伤过程中的铁蛋白沉积症
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-27 DOI: 10.1016/j.biopha.2024.117513
Cardiac arrest (CA) is a significant challenge for emergency physicians worldwide and leads to increased morbidity and mortality rates. The poor prognosis of CA primarily stems from the complexity and irreversibility of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis, a form of programmed cell death characterized by iron overload and lipid peroxidation, plays a crucial role in the progression and treatment of CIRI. In this review, we highlight the mechanisms of ferroptosis within the context of CIRI, focusing on its role as a key contributor to neuronal damage and dysfunction post-CA. We explore the crucial involvement of the nuclear factor erythroid 2-related factor (Nrf2)-mediated signaling pathway in modulating ferroptosis-associated processes during CIRI. Through comprehensive analysis of the regulatory role of Nrf2 in the cellular responses to oxidative stress, we highlight its potential as a therapeutic target for mitigating ferroptotic cell death and improving the neurological prognosis of patients experiencing CA. Furthermore, we discuss interventions targeting the Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element pathway, including the use of traditional Chinese medicine and Western medicine, which demonstrate potential for attenuating ferroptosis and preserving neuronal function in CIRI. Owing to the limitations in the safety, specificity, and effectiveness of Nrf2-targeted drugs, as well as the technical difficulties and ethical constraints in obtaining the results related to the brain pathological examination of patients, most of the studies focusing on Nrf2-related regulation of ferroptosis in CIRI are still in the basic research stage. Overall, this review aims to provide a comprehensive understanding of the mechanisms underlying ferroptosis in CIRI, offering insights into novel therapeutics aimed at enhancing the clinical outcomes of patients with CA.
心脏骤停(CA)是全世界急诊医生面临的重大挑战,会导致发病率和死亡率上升。心脏骤停的预后不良主要源于脑缺血再灌注损伤(CIRI)的复杂性和不可逆性。铁变态反应是一种以铁超载和脂质过氧化为特征的程序性细胞死亡,在 CIRI 的进展和治疗中起着至关重要的作用。在这篇综述中,我们将着重介绍 CIRI 背景下的铁蜕变机制,重点是铁蜕变作为导致脑梗死后神经元损伤和功能障碍的关键因素的作用。我们探讨了核因子红细胞 2 相关因子(Nrf2)介导的信号通路在调控 CIRI 期间铁蜕变相关过程中的关键作用。通过全面分析 Nrf2 在细胞对氧化应激反应中的调控作用,我们强调了 Nrf2 作为治疗靶点的潜力,可减轻铁沉着细胞死亡,改善 CA 患者的神经系统预后。此外,我们还讨论了针对 Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element 通路的干预措施,包括使用传统中药和西药,这些措施在减轻铁凋亡和保护 CIRI 神经元功能方面具有潜力。由于 Nrf2 靶向药物在安全性、特异性和有效性方面的局限性,以及获得与患者脑部病理检查相关结果的技术难度和伦理限制,大多数关注 Nrf2 相关调控 CIRI 中铁细胞减少的研究仍处于基础研究阶段。总之,本综述旨在提供对 CIRI 中铁细胞减少机制的全面理解,为旨在提高 CA 患者临床疗效的新型疗法提供见解。
{"title":"Nuclear factor erythroid 2-related factor-mediated signaling alleviates ferroptosis during cerebral ischemia-reperfusion injury","authors":"","doi":"10.1016/j.biopha.2024.117513","DOIUrl":"10.1016/j.biopha.2024.117513","url":null,"abstract":"<div><div>Cardiac arrest (CA) is a significant challenge for emergency physicians worldwide and leads to increased morbidity and mortality rates. The poor prognosis of CA primarily stems from the complexity and irreversibility of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis, a form of programmed cell death characterized by iron overload and lipid peroxidation, plays a crucial role in the progression and treatment of CIRI. In this review, we highlight the mechanisms of ferroptosis within the context of CIRI, focusing on its role as a key contributor to neuronal damage and dysfunction post-CA. We explore the crucial involvement of the nuclear factor erythroid 2-related factor (Nrf2)-mediated signaling pathway in modulating ferroptosis-associated processes during CIRI. Through comprehensive analysis of the regulatory role of Nrf2 in the cellular responses to oxidative stress, we highlight its potential as a therapeutic target for mitigating ferroptotic cell death and improving the neurological prognosis of patients experiencing CA. Furthermore, we discuss interventions targeting the Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element pathway, including the use of traditional Chinese medicine and Western medicine, which demonstrate potential for attenuating ferroptosis and preserving neuronal function in CIRI. Owing to the limitations in the safety, specificity, and effectiveness of Nrf2-targeted drugs, as well as the technical difficulties and ethical constraints in obtaining the results related to the brain pathological examination of patients, most of the studies focusing on Nrf2-related regulation of ferroptosis in CIRI are still in the basic research stage. Overall, this review aims to provide a comprehensive understanding of the mechanisms underlying ferroptosis in CIRI, offering insights into novel therapeutics aimed at enhancing the clinical outcomes of patients with CA.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142326480","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of Vitamin D on tumor cell proliferation and migration, tumor initiation and anti-tumor immune response in head and neck squamous cell carcinomas 维生素 D 对头颈部鳞状细胞癌中肿瘤细胞增殖和迁移、肿瘤诱发和抗肿瘤免疫反应的影响
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-27 DOI: 10.1016/j.biopha.2024.117497

Background

Head and neck squamous cell carcinomas (HNSCCs) are among the six most common cancers, with a constantly poor prognosis. Vitamin D has been found to have antineoplastic and immunomodulatory properties in various cancers. This study investigated the impact of Vitamin D on the initiation and progression as well as antitumor immune response in HNSCCs, both in vitro and in vivo.

Methods

An immunocompetent, orthotopic oral carcinogenesis mouse model was used to examine the influence of Vitamin D3 substitution on HNSCC initiation and progression in vivo. Tumor immune infiltration was analyzed by immunohistochemistry targeting CD3, CD8, NKR-P1C, FOXP3, and CD163. Two HPV- and two HPV+ HNSCC cell lines were treated with 1,25-dihydroxyvitamin D3 to analyze effects on tumor cell proliferation, migration and transcriptomic changes using RNA-sequencing, differential gene expression and gene set enrichment analysis.

Results

Vitamin D3 treatment led to a significant suppression of HNSCC initiation and progression, while also stimulating tumor immune infiltration with CD3+, CD8+ and NKR-P1C+ cells and lowering levels of M2 macrophages and Treg cells in vivo. In vitro experiments showed an inhibition of HNSCC cell proliferation and migration in HPV+ and HPV- cell lines. RNA-sequencing showed significant regulations in IL6 JAK STAT3, hypoxia signaling and immunomodulatory pathways upon Vitamin D3 treatment.

Conclusion

The findings of our study highlight the promising potential of Vitamin D in the therapeutic repertoire for HNSCC patients given its immune modulating, anti-proliferative and anti-migratory properties. Clinical transferability of those in vitro and in vivo effects should be further validated in clinical trials.
背景头颈部鳞状细胞癌(HNSCC)是六种最常见的癌症之一,预后一直很差。研究发现,维生素 D 在多种癌症中具有抗肿瘤和免疫调节作用。本研究探讨了维生素 D 在体外和体内对 HNSCC 的发生、发展和抗肿瘤免疫反应的影响。方法采用免疫功能健全的正位口腔癌小鼠模型,研究维生素 D3 替代物在体内对 HNSCC 发生和发展的影响。通过针对 CD3、CD8、NKR-P1C、FOXP3 和 CD163 的免疫组化分析了肿瘤免疫浸润。用 1,25-二羟维生素 D3 处理两种 HPV- 型和两种 HPV+ 型 HNSCC 细胞系,分析其对肿瘤细胞增殖、迁移和转录组变化的影响(采用 RNA 测序、差异基因表达和基因组富集分析)。结果维生素 D3 治疗显著抑制了 HNSCC 的发生和发展,同时还刺激了 CD3+、CD8+ 和 NKR-P1C+ 细胞对肿瘤的免疫浸润,降低了体内 M2 巨噬细胞和 Treg 细胞的水平。体外实验显示,HPV+ 和 HPV- 细胞系的 HNSCC 细胞增殖和迁移受到抑制。RNA 序列分析表明,维生素 D3 治疗后,IL6 JAK STAT3、缺氧信号传导和免疫调节通路都有明显的改变。这些体外和体内效应的临床可转移性应在临床试验中进一步验证。
{"title":"Effects of Vitamin D on tumor cell proliferation and migration, tumor initiation and anti-tumor immune response in head and neck squamous cell carcinomas","authors":"","doi":"10.1016/j.biopha.2024.117497","DOIUrl":"10.1016/j.biopha.2024.117497","url":null,"abstract":"<div><h3>Background</h3><div>Head and neck squamous cell carcinomas (HNSCCs) are among the six most common cancers, with a constantly poor prognosis. Vitamin D has been found to have antineoplastic and immunomodulatory properties in various cancers. This study investigated the impact of Vitamin D on the initiation and progression as well as antitumor immune response in HNSCCs, both <em>in vitro</em> and <em>in vivo</em>.</div></div><div><h3>Methods</h3><div>An immunocompetent, orthotopic oral carcinogenesis mouse model was used to examine the influence of Vitamin D<sub>3</sub> substitution on HNSCC initiation and progression <em>in vivo</em>. Tumor immune infiltration was analyzed by immunohistochemistry targeting CD3, CD8, NKR-P1C, FOXP3, and CD163. Two HPV- and two HPV+ HNSCC cell lines were treated with 1,25-dihydroxyvitamin D<sub>3</sub> to analyze effects on tumor cell proliferation, migration and transcriptomic changes using RNA-sequencing, differential gene expression and gene set enrichment analysis.</div></div><div><h3>Results</h3><div>Vitamin D<sub>3</sub> treatment led to a significant suppression of HNSCC initiation and progression, while also stimulating tumor immune infiltration with CD3+, CD8+ and NKR-P1C+ cells and lowering levels of M2 macrophages and T<sub>reg</sub> cells <em>in vivo</em>. <em>In vitro</em> experiments showed an inhibition of HNSCC cell proliferation and migration in HPV+ and HPV- cell lines. RNA-sequencing showed significant regulations in IL6 JAK STAT3, hypoxia signaling and immunomodulatory pathways upon Vitamin D<sub>3</sub> treatment<strong>.</strong></div></div><div><h3>Conclusion</h3><div>The findings of our study highlight the promising potential of Vitamin D in the therapeutic repertoire for HNSCC patients given its immune modulating, anti-proliferative and anti-migratory properties. Clinical transferability of those <em>in vitro</em> and <em>in vivo</em> effects should be further validated in clinical trials.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142326358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment 优化 CRISPR/Cas9 精确性:减轻脱靶效应,在癌症治疗中安全整合光动力疗法和干细胞疗法
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117516
CRISPR/Cas9 precision genome editing has revolutionized cancer treatment by introducing specific alterations to the cancer genome. But the therapeutic potential of CRISPR/Cas9 is limited by off-target effects, which can cause undesired changes to genomic regions and create major safety concerns. The primary emphasis lies in their implications within the realm of cancer photodynamic therapy (PDT), where precision is paramount. PDT is a promising cancer treatment method; nevertheless, its effectiveness is severely limited and readily leads to recurrence due to the therapeutic resistance of cancer stem cells (CSCs). With a focus on targeted genome editing into cancer cells during PDT and stem cell treatment (SCT), the review aims to further the ongoing search for safer and more accurate CRISPR/Cas9-mediated methods. At the core of this exploration are recent advancements and novel techniques that offer promise in mitigating the risks associated with off-target effects. With a focus on cancer PDT and SCT, this review critically assesses the landscape of off-target effects in CRISPR/Cas9 applications, offering a comprehensive knowledge of their nature and prevalence. A key component of the review is the assessment of cutting-edge delivery methods, such as technologies based on nanoparticles (NPs), to optimize the distribution of CRISPR components. Additionally, the study delves into the intricacies of guide RNA design, focusing on advancements that bolster specificity and minimize off-target effects, crucial elements in ensuring the precision required for effective cancer PDT and SCT. By synthesizing insights from various methodologies, including the exploration of innovative genome editing tools and leveraging robust validation methods and bioinformatics tools, the review aspires to chart a course towards more reliable and precise CRISPR-Cas9 applications in cancer PDT and SCT. For safe PDT and SCT integration in cancer therapy, CRISPR/Cas9 precision optimization is essential. Utilizing sophisticated molecular and computational techniques to address off-target effects is crucial to realizing the therapeutic promise of these technologies, which will ultimately lead to the development of individualized and successful cancer treatment strategies. Our long-term goals are to improve precision genome editing for more potent cancer therapy approaches by refining the way CRISPR/Cas9 is integrated with photodynamic and stem cell therapies.
CRISPR/Cas9 精确基因组编辑技术通过对癌症基因组引入特定的改变,为癌症治疗带来了革命性的变化。但是,CRISPR/Cas9 的治疗潜力受到脱靶效应的限制,脱靶效应可能导致基因组区域发生不希望发生的变化,并带来重大的安全问题。主要重点在于它们对癌症光动力疗法(PDT)领域的影响,在这一领域,精确性是至关重要的。光动力疗法是一种很有前景的癌症治疗方法;然而,由于癌症干细胞(CSCs)的抗药性,其疗效受到严重限制,并很容易导致复发。本综述的重点是在PDT和干细胞治疗(SCT)过程中对癌细胞进行靶向基因组编辑,旨在进一步探索更安全、更精确的CRISPR/Cas9介导方法。这一探索的核心是最近取得的进展和新型技术,它们有望减轻与脱靶效应相关的风险。这篇综述以癌症光动力疗法和造血干细胞移植为重点,批判性地评估了 CRISPR/Cas9 应用中的脱靶效应,提供了有关其性质和普遍性的全面知识。该综述的一个关键部分是评估尖端的递送方法,如基于纳米颗粒(NPs)的技术,以优化 CRISPR 成分的分布。此外,该研究还深入探讨了引导 RNA 设计的复杂性,重点关注提高特异性和最大限度减少脱靶效应的进展,这些都是确保有效的癌症局部放疗和 SCT 所需的精确性的关键因素。通过综合各种方法的见解,包括对创新基因组编辑工具的探索,以及利用可靠的验证方法和生物信息学工具,本综述希望为 CRISPR-Cas9 在癌症局部放疗和 SCT 中更可靠、更精确的应用指明方向。要在癌症治疗中安全整合光动力疗法和 SCT,CRISPR/Cas9 的精确优化至关重要。利用复杂的分子和计算技术来解决脱靶效应,对于实现这些技术的治疗前景至关重要,这将最终促成个体化和成功的癌症治疗策略的开发。我们的长期目标是通过改进CRISPR/Cas9与光动力疗法和干细胞疗法的结合方式,提高基因组编辑的精确度,从而开发出更有效的癌症治疗方法。
{"title":"Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment","authors":"","doi":"10.1016/j.biopha.2024.117516","DOIUrl":"10.1016/j.biopha.2024.117516","url":null,"abstract":"<div><div>CRISPR/Cas9 precision genome editing has revolutionized cancer treatment by introducing specific alterations to the cancer genome. But the therapeutic potential of CRISPR/Cas9 is limited by off-target effects, which can cause undesired changes to genomic regions and create major safety concerns. The primary emphasis lies in their implications within the realm of cancer photodynamic therapy (PDT), where precision is paramount. PDT is a promising cancer treatment method; nevertheless, its effectiveness is severely limited and readily leads to recurrence due to the therapeutic resistance of cancer stem cells (CSCs). With a focus on targeted genome editing into cancer cells during PDT and stem cell treatment (SCT), the review aims to further the ongoing search for safer and more accurate CRISPR/Cas9-mediated methods. At the core of this exploration are recent advancements and novel techniques that offer promise in mitigating the risks associated with off-target effects. With a focus on cancer PDT and SCT, this review critically assesses the landscape of off-target effects in CRISPR/Cas9 applications, offering a comprehensive knowledge of their nature and prevalence. A key component of the review is the assessment of cutting-edge delivery methods, such as technologies based on nanoparticles (NPs), to optimize the distribution of CRISPR components. Additionally, the study delves into the intricacies of guide RNA design, focusing on advancements that bolster specificity and minimize off-target effects, crucial elements in ensuring the precision required for effective cancer PDT and SCT. By synthesizing insights from various methodologies, including the exploration of innovative genome editing tools and leveraging robust validation methods and bioinformatics tools, the review aspires to chart a course towards more reliable and precise CRISPR-Cas9 applications in cancer PDT and SCT. For safe PDT and SCT integration in cancer therapy, CRISPR/Cas9 precision optimization is essential. Utilizing sophisticated molecular and computational techniques to address off-target effects is crucial to realizing the therapeutic promise of these technologies, which will ultimately lead to the development of individualized and successful cancer treatment strategies. Our long-term goals are to improve precision genome editing for more potent cancer therapy approaches by refining the way CRISPR/Cas9 is integrated with photodynamic and stem cell therapies.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142322037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PSF-lncRNA interaction as a target for novel targeted anticancer therapies PSF-lncRNA 相互作用是新型靶向抗癌疗法的目标
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117491
The Polypyrimidine Tract-Binding Protein-Associated Splicing Factor (PSF), a component of the Drosophila Behavior/Human Splicing (DBHS) complex, plays a pivotal role in cancer pathogenesis. The epigenetic regulation mediated by PSF and long noncoding RNA (lncRNA), along with PSF's alternative splicing activity, has been implicated in promoting cancer cell proliferation, migration, invasion, metastasis, and drug resistance in various human cancers. Recent research highlights the therapeutic promise of targeting the PSF-lncRNA interaction to combat aggressive malignancies, making it a compelling target for cancer therapy. This review offers a detailed synthesis of the current understanding of PSF's role in oncogenic pathways and recent progress in identifying inhibitors of PSF-lncRNA interactions. Furthermore, it discusses the potential of using these inhibitors in cancer treatment strategies, especially as adjuncts to immune checkpoint blockade therapies to improve the efficacy of anti-PD-(L)1 treatments in Glioblastoma Multiforme (GBM). By outlining the interaction patterns of existing PSF-lncRNA inhibitors, this article aims to guide the development and refinement of future pharmacological interventions.
多嘧啶簇结合蛋白相关剪接因子(PSF)是果蝇行为/人类剪接(DBHS)复合体的一个组成部分,在癌症发病机制中起着关键作用。PSF 和长非编码 RNA(lncRNA)介导的表观遗传调控,以及 PSF 的替代剪接活性,已被证实与促进各种人类癌症的癌细胞增殖、迁移、侵袭、转移和耐药性有关。最近的研究强调了针对 PSF-lncRNA 相互作用的治疗前景,以对抗侵袭性恶性肿瘤,使其成为癌症治疗的一个引人注目的靶点。本综述详细综述了目前对 PSF 在致癌通路中作用的理解,以及在确定 PSF-lncRNA 相互作用抑制剂方面的最新进展。此外,它还讨论了在癌症治疗策略中使用这些抑制剂的潜力,尤其是将其作为免疫检查点阻断疗法的辅助手段,以提高多形性胶质母细胞瘤(GBM)中抗-PD-(L)1疗法的疗效。本文概述了现有 PSF-lncRNA 抑制剂的相互作用模式,旨在指导未来药物干预措施的开发和完善。
{"title":"PSF-lncRNA interaction as a target for novel targeted anticancer therapies","authors":"","doi":"10.1016/j.biopha.2024.117491","DOIUrl":"10.1016/j.biopha.2024.117491","url":null,"abstract":"<div><div>The Polypyrimidine Tract-Binding Protein-Associated Splicing Factor (PSF), a component of the Drosophila Behavior/Human Splicing (DBHS) complex, plays a pivotal role in cancer pathogenesis. The epigenetic regulation mediated by PSF and long noncoding RNA (lncRNA), along with PSF's alternative splicing activity, has been implicated in promoting cancer cell proliferation, migration, invasion, metastasis, and drug resistance in various human cancers. Recent research highlights the therapeutic promise of targeting the PSF-lncRNA interaction to combat aggressive malignancies, making it a compelling target for cancer therapy. This review offers a detailed synthesis of the current understanding of PSF's role in oncogenic pathways and recent progress in identifying inhibitors of PSF-lncRNA interactions. Furthermore, it discusses the potential of using these inhibitors in cancer treatment strategies, especially as adjuncts to immune checkpoint blockade therapies to improve the efficacy of anti-PD-(L)1 treatments in Glioblastoma Multiforme (GBM). By outlining the interaction patterns of existing PSF-lncRNA inhibitors, this article aims to guide the development and refinement of future pharmacological interventions.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142320170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways 用于骨重塑的天然化合物:针对成骨细胞和相关信号通路
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117490
In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.
在骨代谢和骨重塑过程中,骨髓间充质干细胞(BM-MSCs)在一定条件下分化为成骨细胞(OBs),从而形成新骨,正常骨重建和病理骨改变与成骨细胞的分化和增殖功能密切相关。造血干细胞的成骨分化涉及多种信号通路,它们各自发挥作用,但又错综复杂地相互联系,形成一个复杂的信号调控网络。与化学合成药物相比,天然化合物的不良反应较少,能优化骨骼健康,更适合长期使用。本文以 OBs 为研究对象,总结了目前与 OBs 分化相关的信号通路的研究进展,并综述了具有潜在抗骨质疏松症作用的化学合成药物调控 OBs 介导的骨形成的分子机制。
{"title":"Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways","authors":"","doi":"10.1016/j.biopha.2024.117490","DOIUrl":"10.1016/j.biopha.2024.117490","url":null,"abstract":"<div><div>In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142322034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prostaglandin E2 signaling through prostaglandin E receptor subtype 2 and Nurr1 induces fibroblast growth factor 23 production 通过前列腺素 E 受体亚型 2 和 Nurr1 发出的前列腺素 E2 信号诱导成纤维细胞生长因子 23 的产生
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117475
Bone cells produce fibroblast growth factor 23 (FGF23), a hormone regulating renal phosphate and vitamin D homeostasis, and a paracrine factor produced in further tissues. Chronic kidney disease and cardiovascular disorders are associated with early elevations of plasma FGF23 levels associated with clinical outcomes. FGF23 production is dependent on many conditions including inflammation. Prostaglandin E2 (PGE2) is a major eicosanoid with a broad role in pain, inflammation, and fever. Moreover, it regulates renal blood flow, renin secretion, natriuresis as well as bone formation through prostaglandin E receptor 2 (EP2). Here, we studied the role of PGE2 and its signaling for the production of FGF23. Osteoblast-like UMR-106 cells were exposed to EP receptor agonists, antagonists or RNAi. Wild type and EP2 knockout mice were treated with stable EP2 agonist misoprostol. Fgf23 or Nurr1 gene expression was determined by quantitative real-time PCR, hormone and further blood parameters by enzyme-linked immunosorbent assay and colorimetric methods. PGE2 and EP2 agonists misoprostol and butaprost enhanced FGF23 production in UMR-106 cells, effects mediated by EP2 and transcription factor Nurr1. A single dose of misoprostol up-regulated bone Fgf23 expression and FGF23 serum levels in wild type mice with subtle effects on parameters of mineral metabolism only. Compared to wild type mice, the FGF23 effect of misoprostol was significantly lower in EP2-deficient mice. To conclude, PGE2 signaling through EP2 and Nurr1 induces FGF23 production. Given the broad physiological and pathophysiological implications of PGE2 signaling, this effect is likely of clinical relevance.
骨细胞产生成纤维细胞生长因子 23 (FGF23),这是一种调节肾磷酸盐和维生素 D 平衡的激素,也是其他组织产生的旁分泌因子。慢性肾病和心血管疾病的早期血浆 FGF23 水平升高与临床结果有关。FGF23 的产生取决于许多条件,包括炎症。前列腺素 E2(PGE2)是一种主要的类二十烷,在疼痛、炎症和发热中发挥着广泛的作用。此外,它还能通过前列腺素 E 受体 2(EP2)调节肾血流量、肾素分泌、利尿以及骨形成。在此,我们研究了 PGE2 及其信号传导对 FGF23 生成的作用。成骨细胞样 UMR-106 细胞暴露于 EP 受体激动剂、拮抗剂或 RNAi。用稳定的 EP2 激动剂米索前列醇处理野生型和 EP2 基因敲除小鼠。Fgf23 或 Nurr1 基因表达采用实时定量 PCR 法,激素和其他血液参数采用酶联免疫吸附法和比色法。PGE2和EP2激动剂米索前列醇和丁前列腺素可促进UMR-106细胞中FGF23的产生,其作用由EP2和转录因子Nurr1介导。单剂量的米索前列醇可上调野生型小鼠的骨Fgf23表达和FGF23血清水平,但只对矿物质代谢参数产生微妙影响。与野生型小鼠相比,EP2缺陷小鼠体内米索前列醇的FGF23效应明显降低。总之,PGE2 信号通过 EP2 和 Nurr1 诱导 FGF23 的产生。鉴于 PGE2 信号传导具有广泛的生理和病理生理学影响,这种效应很可能与临床相关。
{"title":"Prostaglandin E2 signaling through prostaglandin E receptor subtype 2 and Nurr1 induces fibroblast growth factor 23 production","authors":"","doi":"10.1016/j.biopha.2024.117475","DOIUrl":"10.1016/j.biopha.2024.117475","url":null,"abstract":"<div><div>Bone cells produce fibroblast growth factor 23 (FGF23), a hormone regulating renal phosphate and vitamin D homeostasis, and a paracrine factor produced in further tissues. Chronic kidney disease and cardiovascular disorders are associated with early elevations of plasma FGF23 levels associated with clinical outcomes. FGF23 production is dependent on many conditions including inflammation. Prostaglandin E<sub>2</sub> (PGE<sub>2</sub>) is a major eicosanoid with a broad role in pain, inflammation, and fever. Moreover, it regulates renal blood flow, renin secretion, natriuresis as well as bone formation through prostaglandin E receptor 2 (EP2). Here, we studied the role of PGE<sub>2</sub> and its signaling for the production of FGF23. Osteoblast-like UMR-106 cells were exposed to EP receptor agonists, antagonists or RNAi. Wild type and EP2 knockout mice were treated with stable EP2 agonist misoprostol. Fgf23 or Nurr1 gene expression was determined by quantitative real-time PCR, hormone and further blood parameters by enzyme-linked immunosorbent assay and colorimetric methods. PGE<sub>2</sub> and EP2 agonists misoprostol and butaprost enhanced FGF23 production in UMR-106 cells, effects mediated by EP2 and transcription factor Nurr1. A single dose of misoprostol up-regulated bone <em>Fgf23</em> expression and FGF23 serum levels in wild type mice with subtle effects on parameters of mineral metabolism only. Compared to wild type mice, the FGF23 effect of misoprostol was significantly lower in EP2-deficient mice. To conclude, PGE<sub>2</sub> signaling through EP2 and Nurr1 induces FGF23 production. Given the broad physiological and pathophysiological implications of PGE<sub>2</sub> signaling, this effect is likely of clinical relevance.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142320226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanism of synergistic action of colistin with resveratrol and baicalin against mcr-1-positive Escherichia coli 秋水仙素与白藜芦醇和黄芩苷对 mcr-1 阳性大肠杆菌的协同作用机制
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117487
The rising incidence of colistin (COL) resistance poses a significant challenge, undermining the therapeutic efficacy of COL against life-threatening bacterial infections. Therefore, the urgent identification and development of new therapeutics are imperative. It has been proven that combinations of antibiotics and promising non-antibiotic agents could be a potential strategy to combat infections caused by MDR pathogens. Due to various antimicrobial properties, medicinal plants have attracted significant attention, which could be promising adjuvant. In this study, we investigated the synergistic effects of combining COL with resveratrol (RST) and baicalin (BAI) against mcr-1-positive Escherichia coli through antibiotic susceptibility testing, checkerboard method and time-killing assays. The mechanisms of combination treatment were analyzed using SEM, fluorometric assays and transcriptome analysis. The molecular docking assay was conducted to elucidate potential interactions between RST, BAI and the MCR-1 protein. Finally, we assessed the in vivo efficacy of combination against mcr-1-positive Escherichia coli. The results demonstrated that the combination of RST, BAI and COL showed significant synergistic activity both in vitro and in vivo. Further mechanistic study revealed that the combination could increase the membrane-damaging ability of COL, disrupt the homeostasis of proton motive force (PMF), inhibit the activity of efflux pumps and impair ATP supply. The molecular docking revealed that RST and BAI could bind to MCR-1 stably, indicating the combination of RST and BAI may be an effective MCR-1 inhibitor. Our findings demonstrated that the combination of RST and BAI might be potential COL adjuvant, providing an alternative approach to address mcr-1-positive Escherichia coli infections.
可乐定(COL)耐药性发生率的上升带来了巨大挑战,削弱了可乐定对危及生命的细菌感染的疗效。因此,当务之急是确定和开发新的疗法。事实证明,抗生素和有前景的非抗生素制剂的组合可能是抗击 MDR 病原体感染的一种潜在策略。由于药用植物具有多种抗菌特性,因此备受关注,它们可能成为有前景的辅助药物。在这项研究中,我们通过抗生素敏感性测试、棋盘格法和时间杀伤试验,研究了 COL 与白藜芦醇(RST)和黄芩苷(BAI)联合使用对 mcr-1 阳性大肠杆菌的协同作用。利用扫描电镜、荧光测定法和转录组分析法分析了联合处理的机制。我们还进行了分子对接试验,以阐明 RST、BAI 和 MCR-1 蛋白之间潜在的相互作用。最后,我们评估了联合疗法对 mcr-1 阳性大肠杆菌的体内疗效。结果表明,RST、BAI 和 COL 的组合在体外和体内都表现出显著的协同活性。进一步的机理研究发现,RST、BAI 和 COL 的组合能增强 COL 的膜破坏能力,破坏质子动力(PMF)的平衡,抑制外排泵的活性,损害 ATP 的供应。分子对接显示,RST和BAI能与MCR-1稳定结合,表明RST和BAI的组合可能是一种有效的MCR-1抑制剂。我们的研究结果表明,RST 和 BAI 的组合可能是潜在的 COL 佐剂,为解决 mcr-1 阳性大肠杆菌感染提供了另一种方法。
{"title":"Mechanism of synergistic action of colistin with resveratrol and baicalin against mcr-1-positive Escherichia coli","authors":"","doi":"10.1016/j.biopha.2024.117487","DOIUrl":"10.1016/j.biopha.2024.117487","url":null,"abstract":"<div><div>The rising incidence of colistin (COL) resistance poses a significant challenge, undermining the therapeutic efficacy of COL against life-threatening bacterial infections. Therefore, the urgent identification and development of new therapeutics are imperative. It has been proven that combinations of antibiotics and promising non-antibiotic agents could be a potential strategy to combat infections caused by MDR pathogens. Due to various antimicrobial properties, medicinal plants have attracted significant attention, which could be promising adjuvant. In this study, we investigated the synergistic effects of combining COL with resveratrol (RST) and baicalin (BAI) against <em>mcr-1</em>-positive <em>Escherichia coli</em> through antibiotic susceptibility testing, checkerboard method and time-killing assays. The mechanisms of combination treatment were analyzed using SEM, fluorometric assays and transcriptome analysis. The molecular docking assay was conducted to elucidate potential interactions between RST, BAI and the MCR-1 protein. Finally, we assessed the <em>in vivo</em> efficacy of combination against <em>mcr-1</em>-positive <em>Escherichia coli</em>. The results demonstrated that the combination of RST, BAI and COL showed significant synergistic activity both <em>in vitro</em> and <em>in vivo</em>. Further mechanistic study revealed that the combination could increase the membrane-damaging ability of COL, disrupt the homeostasis of proton motive force (PMF), inhibit the activity of efflux pumps and impair ATP supply. The molecular docking revealed that RST and BAI could bind to MCR-1 stably, indicating the combination of RST and BAI may be an effective MCR-1 inhibitor. Our findings demonstrated that the combination of RST and BAI might be potential COL adjuvant, providing an alternative approach to address <em>mcr-1</em>-positive <em>Escherichia coli</em> infections.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142322040","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unveiling the antitumor synergy between pazopanib and metformin on lung cancer through suppressing p-Akt/ NF-κB/ STAT3/ PD-L1 signal pathway 揭示帕唑帕尼和二甲双胍通过抑制 p-Akt/ NF-κB/ STAT3/ PD-L1 信号通路对肺癌的抗肿瘤协同作用
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117468
Pazopanib, an inhibitor of the VEGF receptor tyrosine kinase, has demonstrated significant antitumor effects in lung cancer. However, its application as a standard treatment for this type of cancer is limited by its drug resistance and toxicity. Metformin has the potential to combat lung cancer by modifying the tumor’s immune microenvironment.
In this study, we investigated the potential antitumor effects and the associated underlying molecular mechanisms of the combination of pazopanib and metformin in lung cancer. In vitro studies were conducted using the A549 and H460 lung cancer cell lines, whereas urethane-induced lung cancer-bearing mice were used for in vivo assessments. The urethane-induced mice received oral administration of pazopanib (50 mg/kg) and/or metformin (250 mg/kg) for a duration of 21 days.
The results indicated that the MTT assay demonstrated a combined cytotoxic effect of the pazopanib/metformin combination in H460 and A549 cells, as evidenced by CI and DRI analyses. The observed increase in annexin V levels and the corresponding increase in Caspase-3 activity strongly suggest that this combination induced apoptosis.
Furthermore, the pazopanib/metformin combination significantly inhibited the p-Akt/NF-κB/IL-6/STAT3, HIF1α/VEGF, and TLR2/TGF-β/PD-L1 pathways while also increasing CD8 expression in vivo. Immunohistochemical analysis revealed that these antitumor mechanisms were manifested by the suppression of the proliferation marker Ki67. In conclusion, these findings revealed that metformin augments the antitumor efficacy of pazopanib in lung cancer by simultaneously targeting proliferative, angiogenic, and immunogenic signaling pathways, metformin enhances the antitumor effectiveness of pazopanib in lung cancer, making it a promising therapeutic option for lung cancer.
帕唑帕尼是一种血管内皮生长因子受体酪氨酸激酶抑制剂,已在肺癌中显示出显著的抗肿瘤效果。然而,由于帕唑帕尼的耐药性和毒性,它在肺癌标准治疗中的应用受到了限制。在本研究中,我们研究了帕唑帕尼和二甲双胍联合治疗肺癌的潜在抗肿瘤作用及其相关的分子机制。体外研究使用了A549和H460肺癌细胞系,体内评估则使用了尿烷诱导的肺癌小鼠。结果表明,MTT检测显示帕唑帕尼/二甲双胍联合疗法对H460和A549细胞具有联合细胞毒性作用,CI和DRI分析也证明了这一点。此外,帕唑帕尼/二甲双胍组合还能显著抑制 p-Akt/NF-κB/IL-6/STAT3、HIF1α/VEGF 和 TLR2/TGF-β/PD-L1 通路,同时还能增加体内 CD8 的表达。免疫组化分析表明,这些抗肿瘤机制表现为抑制增殖标志物 Ki67。总之,这些研究结果表明,二甲双胍通过同时靶向增殖、血管生成和免疫原性信号通路,增强了帕唑帕尼对肺癌的抗肿瘤疗效。
{"title":"Unveiling the antitumor synergy between pazopanib and metformin on lung cancer through suppressing p-Akt/ NF-κB/ STAT3/ PD-L1 signal pathway","authors":"","doi":"10.1016/j.biopha.2024.117468","DOIUrl":"10.1016/j.biopha.2024.117468","url":null,"abstract":"<div><div>Pazopanib, an inhibitor of the VEGF receptor tyrosine kinase, has demonstrated significant antitumor effects in lung cancer. However, its application as a standard treatment for this type of cancer is limited by its drug resistance and toxicity. Metformin has the potential to combat lung cancer by modifying the tumor’s immune microenvironment.</div><div>In this study, we investigated the potential antitumor effects and the associated underlying molecular mechanisms of the combination of pazopanib and metformin in lung cancer. In vitro studies were conducted using the A549 and H460 lung cancer cell lines, whereas urethane-induced lung cancer-bearing mice were used for <em>in vivo</em> assessments. The urethane-induced mice received oral administration of pazopanib (50 mg/kg) and/or metformin (250 mg/kg) for a duration of 21 days.</div><div>The results indicated that the MTT assay demonstrated a combined cytotoxic effect of the pazopanib/metformin combination in H460 and A549 cells, as evidenced by CI and DRI analyses. The observed increase in annexin V levels and the corresponding increase in Caspase-3 activity strongly suggest that this combination induced apoptosis.</div><div>Furthermore, the pazopanib/metformin combination significantly inhibited the p-Akt/NF-κB/IL-6/STAT3, HIF1α/VEGF, and TLR2/TGF-β/PD-L1 pathways while also increasing CD8 expression <em>in vivo</em>. Immunohistochemical analysis revealed that these antitumor mechanisms were manifested by the suppression of the proliferation marker Ki67. In conclusion, these findings revealed that metformin augments the antitumor efficacy of pazopanib in lung cancer by simultaneously targeting proliferative, angiogenic, and immunogenic signaling pathways, metformin enhances the antitumor effectiveness of pazopanib in lung cancer, making it a promising therapeutic option for lung cancer.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142322038","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FTH1P8 induces and transmits docetaxel resistance by inhibiting ferroptosis in prostate cancer FTH1P8 通过抑制前列腺癌中的铁氧化酶诱导并传递多西他赛耐药性
IF 6.9 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Pub Date : 2024-09-26 DOI: 10.1016/j.biopha.2024.117472
Overcoming docetaxel resistance remains a significant challenge in the management of prostate cancer. Previous studies have confirmed a link between ferroptosis and the development of docetaxel resistance. This study revealed that docetaxel-resistant prostate cancer cells presented increased FTH1P8 expression compared with docetaxel-sensitive cells. Decreasing the level of FTH1P8 counteracted docetaxel resistance and facilitated docetaxel-induced ferroptosis, which is characterized by an increase in intracellular Fe2+ concentration, lipid peroxidation levels (lipid ROS), reactive oxygen species (ROS) accumulation, malondialdehyde (MDA) production and mitochondrial damage, a decrease in the Fe3+ concentration and glutathione (GSH) content, and the ability to inhibit hydroxyl radical (·OH) and the mitochondrial membrane potential (MMP). Conversely, increasing the level of FTH1P8 had the opposite effect. A positive correlation was revealed between the expression of FTH1P8 and its parental gene FTH1 in prostate cancer tissues in The Cancer Genome Atlas (TCGA) database. Molecular investigations revealed that FTH1P8 expression increased through miR-1252–5p. Furthermore, rescue experiments confirmed that FTH1 mediated the inhibitory effect of FTH1P8 on ferroptosis. Moreover, FTH1P8 was discovered to play a role in the spread of docetaxel resistance via exosomes. Docetaxel-siRNA targeting FTH1P8 (siFTH1P8)-nanoliposomes (DOC-siFTH1P8-LIP), which can codeliver docetaxel and siFTH1P8, significantly inhibited docetaxel resistance in cells. These results indicated that FTH1P8 can function as both an indicator and a treatment target for docetaxel resistance. The use of DOC-siFTH1P8-LIP demonstrated promising therapeutic effects on docetaxel-resistant cells, suggesting a novel option for treating docetaxel-resistant prostate cancer.
克服多西他赛耐药性仍然是治疗前列腺癌的重大挑战。先前的研究证实了铁蛋白沉积与多西他赛耐药之间的联系。本研究发现,与多西他赛敏感细胞相比,多西他赛耐药前列腺癌细胞的FTH1P8表达增加。降低 FTH1P8 的水平可抵消多西他赛耐药性,并促进多西他赛诱导的铁变态反应、活性氧(ROS)积累、丙二醛(MDA)产生和线粒体损伤,Fe3+浓度和谷胱甘肽(GSH)含量降低,以及抑制羟自由基(-OH)和线粒体膜电位(MMP)的能力。相反,提高 FTH1P8 的水平则会产生相反的效果。癌症基因组图谱(TCGA)数据库显示,前列腺癌组织中 FTH1P8 的表达与其亲代基因 FTH1 呈正相关。分子研究发现,FTH1P8 的表达通过 miR-1252-5p 增加。此外,拯救实验证实,FTH1介导了FTH1P8对铁凋亡的抑制作用。此外,研究还发现 FTH1P8 在多西他赛耐药性通过外泌体扩散的过程中发挥作用。多西他赛-siRNA靶向FTH1P8(siFTH1P8)-nanoliposomes(DOC-siFTH1P8-LIP)能编码传递多西他赛和siFTH1P8,能显著抑制细胞对多西他赛的耐药性。这些结果表明,FTH1P8既可以作为多西他赛耐药的指标,也可以作为多西他赛耐药的治疗靶点。使用 DOC-siFTH1P8-LIP 对多西他赛耐药细胞具有良好的治疗效果,为治疗多西他赛耐药前列腺癌提供了一种新的选择。
{"title":"FTH1P8 induces and transmits docetaxel resistance by inhibiting ferroptosis in prostate cancer","authors":"","doi":"10.1016/j.biopha.2024.117472","DOIUrl":"10.1016/j.biopha.2024.117472","url":null,"abstract":"<div><div>Overcoming docetaxel resistance remains a significant challenge in the management of prostate cancer. Previous studies have confirmed a link between ferroptosis and the development of docetaxel resistance. This study revealed that docetaxel-resistant prostate cancer cells presented increased FTH1P8 expression compared with docetaxel-sensitive cells. Decreasing the level of FTH1P8 counteracted docetaxel resistance and facilitated docetaxel-induced ferroptosis, which is characterized by an increase in intracellular Fe<sup>2+</sup> concentration, lipid peroxidation levels (lipid ROS), reactive oxygen species (ROS) accumulation, malondialdehyde (MDA) production and mitochondrial damage, a decrease in the Fe<sup>3+</sup> concentration and glutathione (GSH) content, and the ability to inhibit hydroxyl radical (·OH) and the mitochondrial membrane potential (MMP). Conversely, increasing the level of FTH1P8 had the opposite effect. A positive correlation was revealed between the expression of FTH1P8 and its parental gene FTH1 in prostate cancer tissues in The Cancer Genome Atlas (TCGA) database. Molecular investigations revealed that FTH1P8 expression increased through miR-1252–5p. Furthermore, rescue experiments confirmed that FTH1 mediated the inhibitory effect of FTH1P8 on ferroptosis. Moreover, FTH1P8 was discovered to play a role in the spread of docetaxel resistance via exosomes. Docetaxel-siRNA targeting FTH1P8 (siFTH1P8)-nanoliposomes (DOC-siFTH1P8-LIP), which can codeliver docetaxel and siFTH1P8, significantly inhibited docetaxel resistance in cells. These results indicated that FTH1P8 can function as both an indicator and a treatment target for docetaxel resistance. The use of DOC-siFTH1P8-LIP demonstrated promising therapeutic effects on docetaxel-resistant cells, suggesting a novel option for treating docetaxel-resistant prostate cancer.</div></div>","PeriodicalId":8966,"journal":{"name":"Biomedicine & Pharmacotherapy","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142322039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biomedicine & Pharmacotherapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1