Dual targeting

双重靶向
  • 文章类型: Journal Article
    目的:结直肠癌是一种极具侵袭性的癌症,常导致死亡。乳铁蛋白显示出靶向和治疗结直肠癌的潜力;然而,口腔递送面临阻碍临床应用的障碍。我们设计了双反应性乳铁蛋白纳米结构微珠来克服递送障碍并增强药物靶向性。
    方法:将疏水性药物美沙拉嗪(MSZ)与乳铁蛋白偶联,形成两亲性偶联纳米颗粒,分散在水中。然后将脂溶性多酚药物白藜芦醇(RSV)包封到LF-MSZ纳米颗粒的疏水核中。为了赋予热响应特性,双有效载荷NP与PNIPAAm外壳耦合;最后,为了进一步赋予纳米颗粒胃肠道抗性和pH响应性,纳米颗粒被微囊化到离子交联的果胶-藻酸盐珠中。
    结果:纳米颗粒通过LF受体介导的内吞作用对HCT结肠癌细胞显示出增强的内化和细胞毒性。温度敏感性聚合物赋予了热触发和调节的释放。涂层保护药物免于降解。口服微珠显著降低小鼠结肠癌模型的肿瘤负荷,降低癌胚抗原和提高抗氧化酶。细胞凋亡途径被刺激,由升高的Bax/Bcl2比率和caspase-3/9表达指示。
    结论:总体而言,我们提出创新的乳铁蛋白纳米结构微珠作为口服结直肠癌治疗的范式转变。
    OBJECTIVE: Colorectal cancer is an extremely aggressive form of cancer that often leads to death. Lactoferrin shows potential for targeting and treating colorectal cancer; however, oral delivery faces hurdles hampering clinical applications. We engineered dual-responsive lactoferrin nanostructured microbeads to overcome delivery hurdles and enhance drug targeting.
    METHODS: The hydrophobic drug mesalazine (MSZ) was coupled to lactoferrin to form amphiphilic conjugate nanoparticles, dispersed in water. The lipid-soluble polyphenolic drug resveratrol (RSV) was then encapsulated into the hydrophobic core of LF-MSZ nanoparticles. To impart thermoresponsive properties, the dual-payload NPs were coupled with a PNIPAAm shell; finally, to further endow the nanoparticles with gastrointestinal resistance and pH responsiveness, the nanoparticles were microencapsulated into ionically cross-linked pectin-alginate beads.
    RESULTS: The nanoparticles showed enhanced internalization and cytotoxicity against HCT colon cancer cells via LF-receptor-mediated endocytosis. Thermal triggering and tuned release were conferred by the temperature-sensitive polymer. The coatings protected the drugs from degradation. Orally delivered microbeads significantly reduced tumor burden in a mouse colon cancer model, lowering carcinoembryonic antigen and elevating antioxidant enzymes. Apoptotic pathways were stimulated, indicated by heightened Bax/Bcl2 ratio and caspase-3/9 expression.
    CONCLUSIONS: Overall, we propose the innovative lactoferrin nanostructured microbeads as a paradigm shift in oral colorectal cancer therapeutics.
    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
    这里,开发了一种无分离,无标签的便携式aptasensor,用于快速,灵敏地分析肿瘤衍生的外来体(TEX)。它集成了平行滚环扩增(RCA)反应,金属离子或小分子与核酸特异性构象的选择性结合,和一个低成本的,高灵敏度手持式荧光计。肺癌,例如,在其外泌体上靶向两种典型的生物标志物(粘蛋白1和程序性细胞死亡配体1(PD-L1))。适体对靶标的亲和力调节了RCA产物(T-Hg2+-T和富含胞嘧啶(C)的单链DNA)的量,进而影响量子点(QDs)和亚甲基蓝(MB)的荧光强度。结果表明,手持式荧光计用于细胞衍生的外来体的检测限(LOD)可低至30颗粒mL-1。此外,它的特异性,灵敏度,曲线下面积(AUC)为93%(14/15),92%(23/25),和0.956,如通过对40个临床样品的分析所确定的。用手持式荧光计重新测试这些样品中的16个,在荧光计结果与从临床计算机断层扫描(CT)和病理学获得的结果之间产生了强烈的一致性。
    Here, a separation-free and label-free portable aptasensor is developed for rapid and sensitive analysis of tumor-derived exosomes (TEXs). It integrated a parallel rolling circle amplification (RCA) reaction, selective binding of metal ions or small molecules to nucleic acid-specific conformations, and a low-cost, highly sensitive handheld fluorometer. Lung cancer, for example, is targeted with two typical biomarkers (mucin 1 and programmed cell death ligand 1 (PD-L1)) on its exosomes. The affinity of aptamers to the targets modulated the amount of RCA products (T-Hg2+-T and cytosine (C)-rich single-stranded DNA), which in turn affected the fluorescence intensity of quantum dots (QDs) and methylene blue (MB). The results revealed that the limit of detection (LOD) of the handheld fluorometer for cell-derived exosomes can be as low as 30 particles mL-1. Moreover, its specificity, sensitivity, and area under the curve (AUC) are 93% (14/15), 92% (23/25), and 0.956, as determined by the analysis of 40 clinical samples. Retesting 16 of these samples with the handheld fluorometer yielded strong concordance between the fluorometer results and those acquired from clinical computed tomography (CT) and pathology.
    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
    几乎所有的线粒体蛋白都由核基因编码,并在细胞质中作为前体蛋白合成。这些前体的氨基酸序列中的信号确保它们的靶向和易位到线粒体中。然而,在许多情况下,只有特定蛋白质的一定部分被转运到线粒体中,而其余的要么留在胞质溶胶中,要么经历反向移位到胞质溶胶中,并且可以填充其他细胞区室。这种现象称为双重定位,可以通过不同的机制引起。这些包括替代的开始或停止密码子,差异转录本,和模糊或竞争的靶向序列。在许多情况下,双重定位可以作为减少所需基因数量的经济策略;例如,当线粒体和叶绿体或线粒体和细胞核/细胞质都需要相同的酶组时。这种情况经常使用不明确的靶向序列来在两个细胞器之间分配蛋白质。然而,替代本地化也可以用于信令,例如,当非输入前体充当线粒体自噬信号时,或者当它们代表细胞核中的转录因子以诱导线粒体未折叠应激反应时。这篇综述概述了双重靶向的机制和生理后果。
    Almost all mitochondrial proteins are encoded by nuclear genes and synthesized in the cytosol as precursor proteins. Signals in the amino acid sequence of these precursors ensure their targeting and translocation into mitochondria. However, in many cases, only a certain fraction of a specific protein is transported into mitochondria, while the rest either remains in the cytosol or undergoes reverse translocation to the cytosol, and can populate other cellular compartments. This phenomenon is called dual localization which can be instigated by different mechanisms. These include alternative start or stop codons, differential transcripts, and ambiguous or competing targeting sequences. In many cases, dual localization might serve as an economic strategy to reduce the number of required genes; for example, when the same groups of enzymes are required both in mitochondria and chloroplasts or both in mitochondria and the nucleus/cytoplasm. Such cases frequently employ ambiguous targeting sequences to distribute proteins between both organelles. However, alternative localizations can also be used for signaling, for example when non-imported precursors serve as mitophagy signals or when they represent transcription factors in the nucleus to induce the mitochondrial unfolded stress response. This review provides an overview regarding the mechanisms and the physiological consequences of dual targeting.
    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
    治疗性癌症疫苗有可能诱导已建立的肿瘤消退,根除微小残留病变,防止转移和复发,但它们的功效受到可溶性抗原和促进肿瘤生长的免疫抑制性肿瘤相关巨噬细胞(TAMs)的低抗原性的限制.在这项研究中,我们报告了一种克服这些防御的新策略:双靶向病毒样颗粒(VLP)纳米疫苗(NV),N-M2T-gp100HBcNV,同时具备SIGNR+树突状细胞(DCs)/TAMs靶向能力和肿瘤相关抗原(TAA)的高密度展示能力。基于N-M2T-gp100HBcNVs的免疫疗法已证明肿瘤相关抗原(TAA)与肿瘤微环境的免疫组成之间的最佳相互作用。在黑色素瘤模型中,N-M2T-gp100HBcVLP显着减少原位和外镜肿瘤生长,并提供了长期的免疫保护。这种显著的抗肿瘤效果是通过有效增强T细胞和M2样TAM的再极化实现的。这项工作为开发个性化肿瘤疫苗开辟了令人兴奋的途径,不仅针对黑色素瘤,而且针对基于功能化VLP的广泛癌症类型。本文受版权保护。保留所有权利。
    Therapeutic cancer vaccines have the potential to induce regression of established tumors, eradicate microscopic residual lesions, and prevent metastasis and recurrence, but their efficacy is limited by the low antigenicity of soluble antigens and the immunosuppressive tumor-associated macrophages (TAMs) that promote tumor growth. In this study, a novel strategy is reported for overcoming these defenses: a dual-targeting nano-vaccine (NV) based on hepatitis B core antigen (HBcAg) derived virus-like particles (VLPs), N-M2T-gp100 HBc NV, equipped with both SIGNR+ dendritic cells (DCs)/TAMs-targeting ability and high-density display of tumor-associated antigen (TAA). N-M2T-gp100 HBc NVs-based immunotherapy has demonstrated an optimal interaction between tumor-associated antigens (TAAs) and the immune composition of the tumor microenvironment. In a melanoma model, N-M2T-gp100 HBc VLPs significantly reducing in situ and abscopal tumor growth, and provide long-term immune protection. This remarkable anti-tumor effect is achieved by efficiently boosting of T cells and repolarizing of M2-like TAMs. This work opens exciting avenues for the development of personalized tumor vaccines targeting not just melanoma but potentially a broad range of cancer types based on functionalized VLPs.
    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
    在这项研究中,我们提出了一种结合透明质酸(HA)的新方法,叶酸(FA),和雷公藤红素(CLS)在聚合物胶束系统(HF-CLS/MLs)中,提供针对乳腺癌的双重行动策略。通过薄膜水化法制备聚合物混合胶束,建立了全面的质量控制参数,包括颗粒大小,多分散指数,zeta电位,表面形态,封装效率,药物含量,体外药物释放,和储存稳定性评估。发现CLS-HF/MLs胶束的平均粒径为120nm,其载药量和包封率分别为15.9%和89.52%,分别。体外释放数据显示,与游离药物相比,CLS-HF/MLs靶向混合胶束显示出延长的释放曲线。此外,所开发的聚合物混合胶束的稳定性在粒度和药物含量方面保持长达8周的储存。此外,流式细胞术和共聚焦激光扫描显微镜研究均表明,CLS-HF/MLs混合胶束对MCF-7细胞系的细胞摄取效率和细胞毒性显着增强。在药代动力学分析方面,发现CLS-HF/MLs混合胶束的半衰期和AUC值比游离药物(CLS)的值高大约4.71-和7.36-倍,分别。由于双重受体(CD44和叶酸)靶向作用,CLS-HF/MLs胶束表现出显着的抗肿瘤功效(几乎完全消除了4种带有T1细胞的肿瘤异种移植物小鼠模型),副作用最小。当考虑到我们目前研究的累积结果时,很明显,为化疗设计的混合胶束为乳腺癌的治疗提供了有希望的和潜在有效的治疗途径.
    In this study, we have proposed a novel approach that combines hyaluronic acid (HA), folic acid (FA), and celastrol (CLS) within a polymeric micelle system (CLS-HF/MLs), offering a dual-action strategy against breast cancer. Polymeric mixed micelles were prepared through the thin-film hydration method, and comprehensive quality control parameters were established, encompassing particle size, polydispersity index, zeta potential, surface morphology, encapsulation efficiency, drug content, in vitro drug release, and storage stability assessment. The average particle size of CLS-HF/MLs micelles was found to be 120 nm and their drug loading and encapsulation efficiencies were 15.9 % and 89.52 %, respectively. The in vitro release data showed that the CLS-HF/MLs targeted mixed micelles displayed a prolonged release profile compared to the free drug. Additionally, the stability of the developed polymeric mixed micelles was maintained for up to 8 weeks of storage in terms of particle size and drug content. Furthermore, both flow cytometry and confocal laser scanning microscopy studies indicated a significant enhancement in the cellular uptake efficiency and cytotoxicity of CLS-HF/MLs mixed micelles against MCF-7 cell line. In terms of pharmacokinetic analysis, the half-life and AUC values of CLS-HF/MLs mixed micelles were found to be approximately 4.71- and 7.36-folds higher than the values of free drug (CLS), respectively. The CLS-HF/MLs micelles exhibited remarkable antitumor efficacy (almost complete ablation of the 4 T1-cell bearing tumor xenografts mouse model) due to the dual receptor (CD44 and folate) targeting effects with minimal side effects. When considering the cumulative findings of our present research, it becomes evident that mixed micelles designed for chemotherapy offer a promising and potentially effective therapeutic avenue for the treatment of breast 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
    动脉粥样硬化(AS),心血管疾病的病理原因,内皮损伤的结果,局部进行性炎症,和过度的脂质积累。富含泡沫细胞的AS斑块容易破裂形成血栓,会导致危及生命的并发症.因此,动脉粥样硬化斑块易损性评估和早期干预对于降低心血管疾病相关死亡率至关重要.在这项工作中,合成了荧光探针FC-TPA,在质子化和非质子化之间转换荧光状态,减少背景荧光和增强信噪比。在此基础上,将FC-TPA加载到磷脂酰丝氨酸靶向肽(PTP)修饰的环糊精(CD)中,并用透明质酸(HA)包被,以构建智能响应性诊断纳米平台(HA@PCFT)。HA@PCFT有效靶向动脉粥样硬化斑块,利用双重瞄准机制。HA与CD44强烈结合,而PTP与磷脂酰丝氨酸结合,使纳米颗粒聚集在病变部位。ROS充当探针的智能释放开关。体外和体内评估均证实了HA@PCFT纳米颗粒(NP)的令人印象深刻的脂质特异性荧光成像能力。通过荧光成像检测动脉粥样硬化斑块中的脂质负荷将有助于评估动脉粥样硬化斑块的脆弱性。重要声明:目前,已经开发了许多荧光探针用于脂质成像。然而,一些挑战,包括水溶性不足,非特异性分布模式,和荧光背景干扰,极大地限制了它们在体内的进一步应用。为了克服这些限制,已经设计和合成了一个荧光分子,通过理论和实验方法深入研究其光物理性质。有趣的是,这种荧光分子表现出可逆的荧光转换能力,由氢键介导,有效减轻背景荧光干扰。此外,荧光分子已经成功地加载到纳米载体功能化的主动靶向能力,显着提高了荧光分子的溶解度,并减少了它们在体内的非特异性分布,可用于动脉粥样硬化的有效目标成像。本研究为评价此类荧光染料的性能提供了有价值的参考,并为动脉粥样硬化的靶向递送系统的设计提供了有希望的观点。
    Atherosclerosis (AS), a pathological cause of cardiovascular disease, results from endothelial injury, local progressive inflammation, and excessive lipid accumulation. AS plaques rich in foam cells are prone to rupture and form thrombus, which can cause life-threatening complications. Therefore, the assessment of atherosclerotic plaque vulnerability and early intervention are crucial in reducing the mortality rates associated with cardiovascular disease. In this work, A fluorescent probe FC-TPA was synthesized, which switches the fluorescence state between protonated and non-protonated, reducing background fluorescence and enhancing imaging signal-to-noise ratio. On this basis, FC-TPA is loaded into cyclodextrin (CD) modified with phosphatidylserine targeting peptide (PTP) and coated with hyaluronic acid (HA) to construct the intelligent responsive diagnostic nanoplatform (HA@PCFT). HA@PCFT effectively targets atherosclerotic plaques, utilizing dual targeting mechanisms. HA binds strongly to CD44, while PTP binds to phosphatidylserine, enabling nanoparticle aggregation at the lesion site. ROS acts as a smart release switch for probes. Both in vitro and in vivo evaluations confirm impressive lipid-specific fluorescence imaging capabilities of HA@PCFT nanoparticles (NPs). The detection of lipid load in atherosclerotic plaque by fluorescence imaging will aid in assessing the vulnerability of atherosclerotic plaque. STATEMENT OF SIGNIFICANCE: Currently, numerous fluorescent probes have been developed for lipid imaging. However, some challenges including inadequate water solubility, nonspecific distribution patterns, and fluorescence background interference, have greatly limited their further applications in vivo. To overcome these limitations, a fluorescent molecule has been designed and synthesized, thoroughly investigating its photophysical properties through both theoretical and experimental approaches. Interestingly, this fluorescent molecule exhibits the reversible fluorescence switching capabilities, mediated by hydrogen bonds, which effectively mitigate background fluorescence interference. Additionally, the fluorescent molecules has been successfully loaded into nanocarriers functionalized with the active targeting abilities, which has significantly improved the solubility of the fluorescent molecules and reduced their nonspecific distribution in vivo for an efficient target imaging in atherosclerosis. This study provides a valuable reference for evaluating the performance of such fluorescent dyes, and offers a promising perspective on the design of the target delivery systems for atherosclerosis.
    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
    目前对多发性硬化症(MS)的药理学方法靶向其炎症和自身免疫成分,但促进髓鞘再生和轴突修复的有效治疗方法仍然缺乏。因此,我们选择了已知与MS发病机理有关的两个靶标:N-酰基乙醇胺水解酸酰胺酶(NAAA)和糖原合酶激酶3β(GSK-3β)。我们测试了抑制这些靶标是否对实验性自身免疫性脑脊髓炎(EAE)具有治疗作用,MS的动物模型两种选定的小分子化合物对NAAA和GSK-3β的联合抑制作用,ARN16186(NAAA抑制剂)和AF3581(GSK-3β抑制剂),有效缓解疾病进展,将动物从瘫痪中拯救出来,防止病理恶化。两种抑制剂的互补活性减少了免疫细胞向脊髓的浸润,并导致脱髓鞘后在轴突周围形成薄的髓鞘。具体来说,抑制NAAA和GSK-3β调节了受EAE影响的小鼠中NF-kB和STAT3转录因子的过度激活,并诱导了β-catenin的核易位,减少炎症损伤和促进髓鞘再生过程。总的来说,这项工作表明,对MS进展的关键方面进行双重靶向可能是解决病理学的创新药理学方法。
    The current pharmacological approaches to multiple sclerosis (MS) target its inflammatory and autoimmune components, but effective treatments to foster remyelination and axonal repair are still lacking. We therefore selected two targets known to be involved in MS pathogenesis: N-acylethanolamine-hydrolyzing acid amidase (NAAA) and glycogen synthase kinase-3β (GSK-3β). We tested whether inhibiting these targets exerted a therapeutic effect against experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The combined inhibition of NAAA and GSK-3β by two selected small-molecule compounds, ARN16186 (an NAAA inhibitor) and AF3581 (a GSK-3β inhibitor), effectively mitigated disease progression, rescuing the animals from paralysis and preventing a worsening of the pathology. The complementary activity of the two inhibitors reduced the infiltration of immune cells into the spinal cord and led to the formation of thin myelin sheaths around the axons post-demyelination. Specifically, the inhibition of NAAA and GSK-3β modulated the over-activation of NF-kB and STAT3 transcription factors in the EAE-affected mice and induced the nuclear translocation of β-catenin, reducing the inflammatory insult and promoting the remyelination process. Overall, this work demonstrates that the dual-targeting of key aspects responsible for MS progression could be an innovative pharmacological approach to tackle the pathology.
    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
    背景:在各种癌症中通常观察到组蛋白脱乙酰酶(HDAC)和核糖核苷酸还原酶(RR)酶的异常表达。研究人员正在癌症研究中关注这些酶,目的是开发有效的化疗药物来治疗癌症。与单一疗法相比,用双重HDAC/RR抑制剂同时靶向HDAC和RR在癌症治疗中显示出增强的有效性。使其成为一个有前途的战略。
    目的:本研究的目的是合成和评估1,10-菲咯啉基羟肟酸盐衍生物的抗癌特性,将其表征为一种新型的双重HDAC/RR抑制剂。
    方法:N1-羟基-N8-(1,10-菲罗啉-5-基)辛二酰胺(PA),一种1,10-菲咯啉基羟肟酸盐衍生物,合成和结构表征。对该化合物进行了抗癌的体外评估,HDAC,和RR抑制活性。进一步研究了计算机对接和分子动力学模拟,以探索其与HDAC和RRM2的相互作用。
    结果:结构证实的PA在SiHa细胞中表现出抗增殖活性,IC50为16.43μM。它对HDAC和RR显示出有效的抑制活性,IC50值为10.80μM和9.34μM,分别。HDAC和RR的共同抑制导致SiHa细胞凋亡诱导的细胞死亡,由活性氧(ROS)的积累介导。计算机对接研究表明,PA可以有效地结合HDAC同工型和RRM2的活性位点。此外,PA表现出与HDAC7更有利的相互作用,显示-9.633kcal/mol的对接评分,与标准HDAC抑制剂辛二酰苯胺异羟肟酸(SAHA)相比,对HDAC7的对接评分为-8.244kcal/mol。
    结论:本研究强调了设计潜在的1,10-菲咯啉异羟肟酸衍生物作为新型的双重HDAC和RR抑制抗癌分子的前景。
    BACKGROUND: Aberrant expression of histone deacetylases (HDACs) and ribonucleotide reductase (RR) enzymes are commonly observed in various cancers. Researchers are focusing on these enzymes in cancer studies with the aim of developing effective chemotherapeutic drugs for cancer treatment. Targeting both HDAC and RR simultaneously with a dual HDAC/RR inhibitor has exhibited enhanced effectiveness compared to monotherapy in cancer treatment, making it a promising strategy.
    OBJECTIVE: The objective of the study is to synthesize and assess the anti-cancer properties of a 1,10-phenanthroline-based hydroxamate derivative, characterizing it as a novel dual HDAC/RR inhibitor.
    METHODS: The N1-hydroxy-N8-(1,10-phenanthrolin-5-yl)octanediamide (PA), a 1,10-phenanthroline-based hydroxamate derivative, was synthesized and structurally characterized. The compound was subjected to in vitro assessments of its anti-cancer, HDAC, and RR inhibitory activities. In silico docking and molecular dynamics simulations were further studied to explore its interactions with HDACs and RRM2.
    RESULTS: The structurally confirmed PA exhibited antiproliferative activity in SiHa cells with an IC50 of 16.43 μM. It displayed potent inhibitory activity against HDAC and RR with IC50 values of 10.80 μM and 9.34 μM, respectively. Co-inhibition of HDAC and RR resulted in apoptosis-induced cell death in SiHa cells, mediated by the accumulation of reactive oxygen species (ROS). In silico docking studies demonstrated that PA can effectively bind to the active sites of HDAC isoforms and RRM2. Furthermore, PA demonstrated a more favorable interaction with HDAC7, displaying a docking score of -9.633 kcal/mol, as compared to the standard HDAC inhibitor suberoylanilide hydroxamic acid (SAHA), which exhibited a docking score of -8.244 kcal/mol against HDAC7.
    CONCLUSIONS: The present study emphasizes the prospect of designing a potential 1,10-phenanthroline hydroxamic acid derivative as a novel dual HDAC and RR-inhibiting anti-cancer molecule.
    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
    冠状病毒一直在畜牧业和公共卫生领域引起全球关注。然而,目前缺乏具有广谱抗病毒活性的有效药物来应对新出现的突变株或耐药性带来的挑战.此外,鉴定多靶点药物的方法也不足。氨肽酶N(APN)和3C样蛋白酶(3CLpro)代表了宿主定向和病毒定向策略的有希望的靶标,分别,开发针对各种冠状病毒的有效药物。在这项研究中,通过靶向这两种蛋白质,马德霉素铵表现出广谱抗病毒作用。来自两个靶蛋白配体的结合域4具有结构相似性,这表明筛选和设计基于这些领域的药物可能表现出广谱和高效的抗病毒活性。此外,已经确定聚醚离子载体携带锌离子的能力可能是它们能够靶向APN并表现出抗病毒作用的原因之一。本实验的发现为未来的药物筛选和设计提供了新的视角,同时也为聚醚离子载体在牲畜健康管理中的利用提供了有价值的参考。
    Coronaviruses have consistently posed a major global concern in the field of livestock industry and public health. However, there is currently a lack of efficient drugs with broad-spectrum antiviral activity to address the challenges presented by emerging mutated strains or drug resistance. Additionally, the method for identifying multitarget drugs is also insufficient. Aminopeptidase N (APN) and 3C-like proteinase (3CLpro) represent promising targets for host-directed and virus-directed strategies, respectively, in the development of effective drugs against various coronaviruses. In this study, maduramycin ammonium demonstrated a broad-spectrum antiviral effect by targeting both of the proteins. The binding domains 4 Å from the ligand of both target proteins shared a structural similarity, suggesting that screening and designing drugs based on these domains might exhibit broad-spectrum and highly effective antiviral activity. Furthermore, it was identified that the polyether ionophores\' ability to carry zinc ion might be one of the reasons why they were able to target APN and exhibit antiviral effect. The findings of this experiment provide novel perspectives for future drug screening and design, while also offering valuable references for the utilization of polyether ionophores in the management of livestock health.
    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
    背景:基于癌症-基质串扰在肿瘤生长中的作用,进展和化学抗性,肿瘤细胞与其基质之间的靶向相互作用提供了新的治疗方法。由于肿瘤微环境的复杂性,选择性作用于肿瘤和基质细胞的双靶向纳米治疗剂可以克服肿瘤细胞靶向单配体纳米药物的局限性。
    方法:嵌入水溶性铱(III)配合物作为光敏剂的金核/二氧化硅壳纳米颗粒和发光探针(Iren-AuSiO2_COOH)被氨基封端的EGFR(CL4)和PDGFRβ(Gint4。T)适体(Iren-AuSiO2_适体)。靶向特异性,和协同光动力和光热效应的单一和双适体修饰的纳米粒子已被评估的共聚焦显微镜和细胞活力测定,分别,在不同的人细胞类型,包括间充质亚型三阴性乳腺癌(MES-TNBC)MDA-MB-231和BT-549细胞系(EGFR和PDGFRβ阳性),管腔/HER2阳性乳腺癌BT-474和表皮样癌A431细胞(仅EGFR阳性)和脂肪间充质基质/干细胞(MSC)(仅PDGFRβ阳性)。缺乏两种受体表达的细胞用作阴性对照。考虑到肿瘤-基质的相互作用,在临床前三维(3D)富含基质的乳腺癌模型中评估荧光成像和细胞毒性。
    结果:我们显示了Iren-AuSiO2_Aptamer纳米平台选择性进入靶细胞的有效能力,杀了他们,通过EGFR和/或PDGFRβ识别。重要的是,通过靶向整个肿瘤中的EGFR+肿瘤/PDGFRβ+基质细胞,在肿瘤细胞和MSCs的3D球体共培养中,双适体工程纳米颗粒比未缀合或单适体缀合纳米颗粒更有效,以及来自病理和分子特征明确的肿瘤的乳腺癌类器官。
    结论:我们的研究提出了智能,新型和安全的多功能纳米平台,同时解决肿瘤微环境中的癌症基质,它们是:(i)通过高度特异性的适体主动递送到靶细胞;(ii)通过它们的发光定位,和(iii)通过微创光激活,发起有效的肿瘤死亡,从而提供创新的精准疗法。鉴于独特的功能,提出的双靶向纳米制剂可能为精准癌症治疗打开一扇新的大门。
    BACKGROUND: Based on the established role of cancer-stroma cross-talk in tumor growth, progression and chemoresistance, targeting interactions between tumor cells and their stroma provides new therapeutic approaches. Dual-targeted nanotherapeutics selectively acting on both tumor and stromal cells may overcome the limits of tumor cell-targeting single-ligand nanomedicine due to the complexity of the tumor microenvironment.
    METHODS: Gold-core/silica-shell nanoparticles embedding a water-soluble iridium(III) complex as photosensitizer and luminescent probe (Iren-AuSiO2_COOH) were efficiently decorated with amino-terminated EGFR (CL4) and PDGFRβ (Gint4.T) aptamers (Iren-AuSiO2_Aptamer). The targeting specificity, and the synergistic photodynamic and photothermal effects of either single- and dual-aptamer-decorated nanoparticles have been assessed by confocal microscopy and cell viability assays, respectively, on different human cell types including mesenchymal subtype triple-negative breast cancer (MES-TNBC) MDA-MB-231 and BT-549 cell lines (both EGFR and PDGFRβ positive), luminal/HER2-positive breast cancer BT-474 and epidermoid carcinoma A431 cells (only EGFR positive) and adipose-derived mesenchymal stromal/stem cells (MSCs) (only PDGFRβ positive). Cells lacking expression of both receptors were used as negative controls. To take into account the tumor-stroma interplay, fluorescence imaging and cytotoxicity were evaluated in preclinical three-dimensional (3D) stroma-rich breast cancer models.
    RESULTS: We show efficient capability of Iren-AuSiO2_Aptamer nanoplatforms to selectively enter into target cells, and kill them, through EGFR and/or PDGFRβ recognition. Importantly, by targeting EGFR+ tumor/PDGFRβ+ stromal cells in the entire tumor bulk, the dual-aptamer-engineered nanoparticles resulted more effective than unconjugated or single-aptamer-conjugated nanoparticles in either 3D spheroids cocultures of tumor cells and MSCs, and in breast cancer organoids derived from pathologically and molecularly well-characterized tumors.
    CONCLUSIONS: Our study proposes smart, novel and safe multifunctional nanoplatforms simultaneously addressing cancer-stroma within the tumor microenvironment, which are: (i) actively delivered to the targeted cells through highly specific aptamers; (ii) localized by means of their luminescence, and (iii) activated via minimally invasive light, launching efficient tumor death, thus providing innovative precision therapeutics. Given the unique features, the proposed dual targeted nanoformulations may open a new door to precision cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号