Drug Repurposing

药物再利用
  • 文章类型: Journal Article
    被诊断患有罕见疾病的患者及其家人拼命寻找组织药物发现运动。不同于默认范式的替代模型提供了真正的机会。有,然而,这种模式的发展没有明确的指导方针,这降低了成功率并提高了成本。我们解决了使新的临床前治疗更容易发现的主要挑战,使用罕见的遗传性截瘫作为典型病例。首先,我们讨论必要的专业知识,以及患者的临床和遗传数据。然后,我们重新审视基因治疗,从头药物开发,和药物再利用,讨论其适用性。此外,我们探索了一组推荐的用于致病变异和蛋白质结构预测的计算机工具,虚拟筛选,和实验验证方法,讨论他们的优点和缺点。最后,我们专注于成功的案例应用。
    Patients diagnosed with rare diseases and their and families search desperately to organize drug discovery campaigns. Alternative models that differ from default paradigms offer real opportunities. There are, however, no clear guidelines for the development of such models, which reduces success rates and raises costs. We address the main challenges in making the discovery of new preclinical treatments more accessible, using rare hereditary paraplegia as a paradigmatic case. First, we discuss the necessary expertise, and the patients\' clinical and genetic data. Then, we revisit gene therapy, de novo drug development, and drug repurposing, discussing their applicability. Moreover, we explore a pool of recommended in silico tools for pathogenic variant and protein structure prediction, virtual screening, and experimental validation methods, discussing their strengths and weaknesses. Finally, we focus on successful case applications.
    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
    由于可靶向生物受体的阴性表达,三阴性乳腺癌(TNBC)被认为是最具侵袭性的乳腺癌形式。与转移能力相关的上皮间质转化(EMT)是其关键特征。作为针对TNBC的尝试,纳米技术被用来增强药物再利用的效果。关于这一点,一个组合治疗模块的结构,其中一个方面是重新利用抗组胺药,负载盐酸异丙嗪的PLGA纳米颗粒。合成的纳米粒子尺寸为217nm,并在522nm处发出荧光,使它们也适合于theranostic应用。该模块的第二个特征是常见的组蛋白脱乙酰酶抑制剂,辛二酰苯胺异羟肟酸(SAHA),用作预处理的形式。实验研究证明了有效的细胞内化和显著的先天抗增殖潜力。SAHA的使用使细胞对装载药物的纳米颗粒处理敏感。机制研究表明ROS产生增加,线粒体功能障碍后细胞凋亡。对蛋白质表达的研究还显示,间充质蛋白如波形蛋白减少了1.90倍;而上皮标志物如E-Cadherin增加了1.42倍,因此表明EMT动力学改变。进一步的发现还提供了对SAHA增强靶向模拟TNBC实体瘤的肿瘤球体的益处的更好的了解。因此,这项研究为将纳米疗法与药物再利用相结合的更合理的翻译验证铺平了道路。 .
    Triple-negative breast cancer (TNBC) is considered the most aggressive form of breast cancer owing to the negative expression of targetable bioreceptors. Epithelial to mesenchymal transition (EMT) associated with metastatic abilities is its critical feature. As an attempt to target TNBC, nanotechnology was utilised to augment the effects of drug repurposing. Concerning that, a combination therapeutic module was structured with one of the aspects being a repurposed antihistamine, promethazine hydrochloride loaded PLGA nanoparticles. The as-synthesized nanoparticles were 217 nm in size and fluoresced at 522 nm, rendering them suitable for theranostic applications too. The second feature of the module was a common histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), used as a form of pre-treatment. Experimental studies demonstrated efficient cellular internalisation and significant innate anti-proliferative potential. The use of SAHA sensitised the cells to the drug loaded nanoparticle treatment. Mechanistic studies showed increase in ROS generation, mitochondrial dysfunction followed by apoptosis. Investigations into protein expression also revealed reduction of mesenchymal proteins like vimentin by 1.90-fold; while increase in epithelial marker like E-Cadherin by 1.42-fold, thus indicating an altered EMT dynamics. Further findings also provided better insight into the benefits of SAHA potentiated targeting of tumor spheroids that mimic solid tumors of TNBC. Thus, this study paves the avenue to a more rational translational validation of combining nanotherapeutics with drug repurposing. .
    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
    精准医学通过靶向负责肿瘤发生的发生和进展的特定分子畸变,彻底改变了现代癌症治疗管理。ROS原癌基因1(ROS1)是一种受体酪氨酸激酶(RTK),可以通过各种信号通路诱导肿瘤发生,如细胞增殖,生存,迁移,和转移。它已成为各种癌症类型的有希望的治疗靶标。然而,用于治疗目的的特定ROS1抑制剂的可用性非常有限.探索用于快速有效治疗的再利用药物是一种有用的方法。在这项研究中,我们利用虚拟筛选和分子动力学(MD)模拟的综合方法重新利用现有的ROS1激酶抑制药物.使用3648种FDA批准的药物的精选库,虚拟筛选鉴定出能够与ROS1激酶结构域结合的药物。结果揭示了两个成功,Midostaurin和Alectinib具有良好的结合谱和与ROS1活性位点残基的稳定相互作用。通过200ns的全原子MD模拟对这些命中进行稳定性评估。MD结果显示Midostaurin和Alectinib与ROS1稳定。一起来看,本研究显示了在进一步验证后,选择具有ROS1抑制潜力的Midostaurin和Alectinib用于治疗的合理框架.
    Precision medicine has revolutionized modern cancer therapeutic management by targeting specific molecular aberrations responsible for the onset and progression of tumorigenesis. ROS proto-oncogene 1 (ROS1) is a receptor tyrosine kinase (RTK) that can induce tumorigenesis through various signaling pathways, such as cell proliferation, survival, migration, and metastasis. It has emerged as a promising therapeutic target in various cancer types. However, there is very limited availability of specific ROS1 inhibitors for therapeutic purposes. Exploring repurposed drugs for rapid and effective treatment is a useful approach. In this study, we utilized an integrated approach of virtual screening and molecular dynamics (MD) simulations of repurposing existing drugs for ROS1 kinase inhibition. Using a curated library of 3648 FDA-approved drugs, virtual screening identified drugs capable of binding to ROS1 kinase domain. The results unveil two hits, Midostaurin and Alectinib with favorable binding profiles and stable interactions with the active site residues of ROS1. These hits were subjected to stability assessment through all-atom MD simulations for 200 ns. MD results showed that Midostaurin and Alectinib were stable with ROS1. Taken together, the study showed a rational framework for the selection of repurposed Midostaurin and Alectinib with ROS1 inhibitory potential for therapeutic management after further validation.
    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
    Rett综合征(RTT)和Rett样综合征[即,CDKL5缺乏症(CDD)和FOXG1综合征]代表罕见但影响深远的神经发育障碍(NDD)。与这些疾病相关的症状的严重程度和复杂性,包括认知障碍,运动功能障碍,癫痫发作和其他神经系统特征显着影响患者和家庭的生活质量。尽管正在进行研究以确定潜在的治疗靶点并开发新的治疗方法,目前的治疗选择仍然有限。在这里,探索了药物再利用(DR)作为解决RTT和相关疾病患者未满足的医疗需求的有希望的途径的潜力。将现有药物用于新的治疗目的,DR提出了一个有吸引力的策略,考虑到中枢神经系统(CNS)的复杂性和血脑屏障穿透的挑战,特别适合神经系统疾病。彻底检查了这些综合征中DR努力的现状,在这些条件下,我们将重点放在共享的分子途径和潜在的常见药物靶标上。
    Rett syndrome (RTT) and Rett-like syndromes [i.e., CDKL5 deficiency disorder (CDD) and FOXG1-syndrome] represent rare yet profoundly impactful neurodevelopmental disorders (NDDs). The severity and complexity of symptoms associated with these disorders, including cognitive impairment, motor dysfunction, seizures and other neurological features significantly affect the quality of life of patients and families. Despite ongoing research efforts to identify potential therapeutic targets and develop novel treatments, current therapeutic options remain limited. Here the potential of drug repurposing (DR) as a promising avenue for addressing the unmet medical needs of individuals with RTT and related disorders is explored. Leveraging existing drugs for new therapeutic purposes, DR presents an attractive strategy, particularly suited for neurological disorders given the complexities of the central nervous system (CNS) and the challenges in blood-brain barrier penetration. The current landscape of DR efforts in these syndromes is thoroughly examined, with partiuclar focus on shared molecular pathways and potential common drug targets across these conditions.
    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
    阿尔茨海默病(Alzheimer’sdisease,AD)是最常见的神经退行性疾病,主要表现为记忆力减退和认知障碍。证据表明,抑郁症是AD患者中常见的合并症,重度抑郁症(MDD)被认为是AD的危险因素。与这两种疾病相关的生物程序之间的串扰使得治疗由它们引起的共病状况非常困难。考虑到AD和MDD的共同病理生理机制,抗抑郁剂药物可能具有有益的治疗效果。在这项研究中,我们旨在探索预防和治疗AD和MDD共病的潜在候选药物.首先,我们通过基于网络的算法评估药物靶点与人蛋白-蛋白相互作用网络(PPIN)上MDD相关基因的距离,筛选了治疗MDD的潜在药物.然后,通过分子对接分析这些药物与tau蛋白和Aβ42肽的亲和力,进一步筛选这些药物,以鉴定可能对AD治疗有效的药物.此外,通过分子动力学模拟确定了最稳定的结合模式,并分析候选药物对AD和MDD发病相关基因的调节作用。共检索到506个MDD相关基因,通过基于网络的方法筛选了831种用于MDD治疗的候选药物。分子对接和分子动力学模拟结果表明,双氢麦角胺与tau蛋白的结合亲和力最低,溴隐亭可以与Aβ42肽形成最稳定的结合模式。进一步的分析发现,双氢麦角胺和溴隐亭都可以调节大脑中与AD和/或MDD发病机理有关的基因的表达。两种药物治疗AD和MDD的确切机制,以及他们的合并症,还不清楚,需要进一步探索来评估它们的作用和机制,在体外和体内。这项研究表明,二氢麦角胺和溴隐亭可能是治疗AD和MDD合并症的潜在候选药物。通过抑制Aβ42和tau蛋白的积累和聚集以及调节脑内疾病相关基因的表达来达到治疗效果。
    Alzheimer\'s disease (AD) is the most prevalent neurodegenerative disease that is characterized by memory loss and cognitive impairment. Evidence shows that depression is a common co-occurrence in AD patients, and major depressive disorder (MDD) is considered a risk factor for AD. The crosstalk between the biological procedures related to the two disorders makes it very difficult to treat the comorbid conditions caused by them. Considering the common pathophysiological mechanisms underlying AD and MDD, antidepressant drugs may have beneficial therapeutic effects against their concurrence. In this study, we aimed to explore the potential drug candidates for the prevention and treatment of the comorbidity of AD and MDD. First, we screened the potential drugs for treating MDD by evaluating the distances of drug targets to MDD-related genes on the human protein-protein interaction network (PPIN) via a network-based algorithm. Then, the drugs were further screened to identify those that may be effective for AD treatment by analyzing their affinities with tau protein and Aβ42 peptide via molecular docking. Furthermore, the most stable binding modes were identified via molecular dynamics simulations, and the regulatory effects of drug candidates on genes involved in the pathogenesis of AD and MDD were analyzed. A total of 506 MDD-related genes were retrieved, and 831 drug candidates for MDD treatment were screened via the network-based approach. The results from molecular docking and molecular dynamics simulations indicated dihydroergotamine had the lowest binding affinity with tau protein and bromocriptine could form the most stable binding mode with Aβ42 peptide. Further analyses found that both dihydroergotamine and bromocriptine could regulate the expression of genes involved in the pathogenesis of AD and/or MDD in the brain. The exact mechanisms of the two drugs in treating AD and MDD, as well as their comorbidity, are still unclear, and further exploration is needed to evaluate their roles and mechanisms, both in vitro and in vivo. This study revealed that dihydroergotamine and bromocriptine may be the potential drug candidates for the treatment of the comorbidity of AD and MDD, and the therapeutic effects may be achieved by inhibiting the accumulation and aggregation of Aβ42 and tau protein and regulating the expression of disease-related genes in the brain.
    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
    Splenic tumors are very common in dogs, and canine hemangiosarcoma (HSA) is one of the most important malignant splenic tumors. Surgery followed by chemotherapy (anthracycline-based protocols) is recommended for treating canine HSA; however, patients still do not achieve long-term survival. Therefore, this research aimed to assess vascular endothelial growth factor receptor-2 (VEGFR-2) and platelet-derived growth factor receptor-β (PDGFR-β) gene expression in formalin-fixed tissues, evaluate the quality of mRNA for quantitative polymerase chain reaction (qPCR) analysis and identify drug repositioning candidates based on VEGFR-2 and PDGFR-β. qPCR analysis identified the relative expression of heterogeneous VEGFR-2 and PDGFR-β, with samples showing no transcripts or very low expression and those with higher relative quantification for both genes. We then used immunohistochemistry to correlate the relative quantification of VEGFR-2 and PDGFR-β transcripts with respective higher protein expression to validate our results. In the next step, we evaluated drug repositioning candidates and identified small molecule inhibitors (i.e. sorafenib) and natural compounds (curcumin and resveratrol) with the ability to block VEGFR-2 and PDGFR-β genes. Overall, our results indicated that VEGFR-2 and PDGFR-β expression is highly variable among canine HSA samples and different drugs can block the expression of both genes. Therefore, a personalized approach could be useful for selecting anti-VEGFR-2 and PDGFR-β therapies and both genes are potential candidates for future oncological panels.
    Os tumores esplênicos são muito comuns em cães, e o hemangiosarcoma (HSA) é um dos tumores esplênicos malignos mais importantes em cães. A cirurgia seguida de quimioterapia (protocolos baseados em antraciclinas) é a abordagem terapêutica mais recomendada para o tratamento da HSA canino; no entanto, os pacientes ainda não alcançam longa sobrevida após tratamento. Portanto, esta pesquisa teve como objetivo avaliar a expressão do receptor do fator de crescimento endotelial vascular-2 (VEGFR-2) e do receptor do fator de crescimento derivado de plaquetas-β (PDGFR-β) em tecidos fixados em formalina e identificar candidatos ao reposicionamento de medicamentos baseado na expressão desses genes. A análise qPCR identificou a expressão relativa heterogênea de VEGFR-2 e PDGFR-β, com amostras sem transcritos ou com expressão muito baixa ou amostras com alta quantificação relativa para ambos os genes. Em seguida, foi realizada o exame imuno-histoquímico para correlacionar a quantificação relativa dos transcritos de VEGFR-2 e PDGFR-β com a respectiva maior expressão proteica para validar nossos resultados. Na próxima etapa, avaliamos candidatos ao reposicionamento de medicamentos e identificamos inibidores de moléculas pequenas (ou seja, sorafenibe) e compostos naturais (curcumina e resveratrol) com capacidade de bloquear os genes VEGFR-2 e PDGFR-β. No geral, nossos resultados indicaram que a expressão de VEGFR-2 e PDGFR-β é altamente variável entre amostras caninas de HSA e diferentes drogas podem bloquear a expressão de ambos os genes. Portanto, uma abordagem personalizada poderia ser útil para selecionar terapias anti-VEGFR-2 e PDGFR-β e ambos os genes são potenciais candidatos para futuros painéis oncológicos.
    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
    1型神经纤维瘤病(NF1)是由NF1基因突变引起的遗传性疾病,与各种症状有关。包括良性肿瘤的形成,叫做神经纤维瘤,在神经内。药物治疗目前是有限的。丝裂原激活的蛋白激酶激酶(MEK)抑制剂司美替尼用于丛状神经纤维瘤(PNs)的一部分,但并不总是有效的,并且可能引起副作用。因此,显然需要发现靶向NF1缺陷肿瘤细胞的新药.使用NF1的果蝇细胞模型,我们进行了合成致死筛选以鉴定新的药物靶标。我们鉴定了54个候选基因,通过可变剂量分析作为二次筛选进行验证。可以使用现有药物靶向与五个候选物相关的途径。其中,氯喹(CQ)和巴弗洛霉素A1,已知靶向自噬途径,显示了选择性杀死NF1缺陷果蝇细胞的最大潜力。当进一步研究自噬相关基因时,我们发现,在30个测试基因中,有14个基因与NF1有合成致死相互作用.这14个基因涉及自噬途径的多个方面,并且可以用介导自噬途径的其他药物靶向,虽然CQ是最有效的。自噬抑制剂的致死作用在一组缺乏NF1的人Schwann细胞系中是保守的,强调他们的翻译潜力。在果蝇NF1体内模型和在小鼠中生长的异种移植NF1缺陷肿瘤细胞系中,CQ的作用也得到了保留。CQ治疗比司美替尼治疗更显著地降低了肿瘤生长。此外,CQ和司美替尼联合治疗导致NF1缺陷型细胞活力进一步降低.总之,NF1缺陷型细胞易受自噬途径破坏的影响。该途径代表了治疗NF1相关肿瘤的有希望的靶标,我们确定CQ是治疗NF1肿瘤的候选药物。
    Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutation of the NF1 gene that is associated with various symptoms, including the formation of benign tumors, called neurofibromas, within nerves. Drug treatments are currently limited. The mitogen-activated protein kinase kinase (MEK) inhibitor selumetinib is used for a subset of plexiform neurofibromas (PNs) but is not always effective and can cause side effects. Therefore, there is a clear need to discover new drugs to target NF1-deficient tumor cells. Using a Drosophila cell model of NF1, we performed synthetic lethal screens to identify novel drug targets. We identified 54 gene candidates, which were validated with variable dose analysis as a secondary screen. Pathways associated with five candidates could be targeted using existing drugs. Among these, chloroquine (CQ) and bafilomycin A1, known to target the autophagy pathway, showed the greatest potential for selectively killing NF1-deficient Drosophila cells. When further investigating autophagy-related genes, we found that 14 out of 30 genes tested had a synthetic lethal interaction with NF1. These 14 genes are involved in multiple aspects of the autophagy pathway and can be targeted with additional drugs that mediate the autophagy pathway, although CQ was the most effective. The lethal effect of autophagy inhibitors was conserved in a panel of human NF1-deficient Schwann cell lines, highlighting their translational potential. The effect of CQ was also conserved in a Drosophila NF1 in vivo model and in a xenografted NF1-deficient tumor cell line grown in mice, with CQ treatment resulting in a more significant reduction in tumor growth than selumetinib treatment. Furthermore, combined treatment with CQ and selumetinib resulted in a further reduction in NF1-deficient cell viability. In conclusion, NF1-deficient cells are vulnerable to disruption of the autophagy pathway. This pathway represents a promising target for the treatment of NF1-associated tumors, and we identified CQ as a candidate drug for the treatment of NF1 tumors.
    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
    重新使用FDA批准的药物是从头药物开发的快速且具有成本效益的替代方案。这里,我们确定了与硼替佐米敏感性有关的基因,预测可能受益于硼替佐米治疗的癌症类型,并评估硼替佐米在乳腺癌中的作用机制(BT-474和ZR-75-30),黑色素瘤(A-375),和体外成胶质细胞瘤(A-172)细胞。来自血液癌症的癌细胞系,肾,神经系统,发现皮肤对硼替佐米的敏感性明显高于其他器官系统。体外研究证实,尽管硼替佐米有效抑制了所有四种细胞系中的β5催化位点,细胞周期阻滞仅在G2/M期诱导,24h后A-375和A-172细胞凋亡。基因组和转录组学分析鉴定了与硼替佐米抗性相关的33个基因(例如ALDH18A1、ATAD2)。一起来看,我们确定了预测硼替佐米敏感性的生物标志物和可能受益于硼替佐米治疗的癌症类型.
    Repurposing of FDA-approved drugs is a quick and cost-effective alternative to de novo drug development. Here, we identify genes involved in bortezomib sensitivity, predict cancer types that may benefit from treatment with bortezomib, and evaluate the mechanism-of-action of bortezomib in breast cancer (BT-474 and ZR-75-30), melanoma (A-375), and glioblastoma (A-172) cells in vitro. Cancer cell lines derived from cancers of the blood, kidney, nervous system, and skin were found to be significantly more sensitive to bortezomib than other organ systems. The in vitro studies confirmed that although bortezomib effectively inhibited the β5 catalytic site in all four cell lines, cell cycle arrest was only induced in G2/M phase and apoptosis in A-375 and A-172 after 24h. The genomic and transcriptomic analyses identified 33 genes (e.g. ALDH18A1, ATAD2) associated with bortezomib resistance. Taken together, we identified biomarkers predictive of bortezomib sensitivity and cancer types that might benefit from treatment with bortezomib.
    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
    腺病毒感染,特别是在儿童中,仍然是一个重要的公共卫生问题,没有批准的针对性治疗。青蒿素及其衍生物,以其在疟疾治疗中的应用而闻名,在最近的研究中显示出抗病毒活性。然而,其对抗人类腺病毒(HAdV)的功效仍有待探索。本研究旨在使用细胞系和原代细胞评估青蒿素及其衍生物对HAdV感染的体外活性。我们的数据表明,青蒿素在很宽的浓度范围内表现出剂量依赖性的抗HAdV活性,没有明显的细胞毒性。机械上,青蒿素不影响病毒附着或进入靶细胞,病毒基因组也不能进入细胞核。相反,它通过抑制病毒DNA复制来抑制HAdV。与其衍生物的比较分析,青蒿琥酯和青蒿素,表现出明显的细胞毒性和抗腺病毒谱,与青蒿素显示出较好的疗效和较低的毒性。使用初级气道上皮细胞模型的进一步验证证实了青蒿素和青蒿素对不同病毒株的抗腺病毒活性。一起,我们的研究结果表明,青蒿素及其衍生物可能是抗HAdV治疗的有希望的候选药物.
    Adenoviral infections, particularly in children, remain a significant public health issue with no approved targeted treatments. Artemisinin and its derivatives, well-known for their use in malaria treatment, have shown antiviral activities in recent studies. However, their efficacy against human adenovirus (HAdV) remains unexplored. This study aimed to assess the activity of artemisinin and its derivatives against HAdV infection in vitro using cell lines and primary cells. Our data revealed that artemisinin exhibited dose-dependent anti-HAdV activity with no apparent cytotoxicity over a wide concentration range. Mechanistically, artemisinin did not affect viral attachment or entry into target cells, nor the viral genome entry into cell nucleus. Instead, it inhibited HAdV through suppression of viral DNA replication. Comparative analysis with its derivatives, artesunate and artemisone, showed distinct cytotoxicity and anti-adenoviral profiles, with artemisone showing superior efficacy and lower toxicity. Further validation using a primary airway epithelial cell model confirmed the anti-adenoviral activity of both artemisinin and artemisone against different virus strains. Together, our findings suggest that artemisinin and its derivatives may be promising candidates for anti-HAdV 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
    每年报告约150万例因真菌感染死亡的病例。对全球健康构成巨大威胁。然而,目前抗真菌药物治疗侵袭性真菌感染的有效性有限.重新使用现有的抗真菌药物是增强其有效性的明智替代方法。这项研究评估了抗病毒药物阿糖腺苷对白色念珠菌ATCC90028的抗真菌功效。通过微量肉汤稀释测定进行抗真菌药敏试验,并进一步处理以找到最低杀真菌浓度。研究阿糖腺苷对白色念珠菌的可能作用方式。活性氧(ROS)积累,核缩合,和凋亡测定。结果表明,白色念珠菌对阿糖腺苷作用敏感,并在150µg/ml时显示出最低抑制浓度。浓度为300微克/毫升,阿糖腺苷对白色念珠菌具有杀真菌活性。300μg/ml阿糖腺苷处理的白色念珠菌细胞表现出减少91%的麦角甾醇含量。膜联蛋白/FITC/PI分析显示,阿糖腺苷(150µg/ml)使晚期凋亡细胞增加了31%。根据分数抑制浓度指数,阿糖腺苷与氟康唑和卡泊芬净对该真菌具有协同作用。在细胞内水平上评估了阿糖腺苷的杀真菌作用的潜在机制,可能是因为核子凝结的增加,增强ROS生成,和细胞周期停滞。总之,这一数据是第一个报道,阿糖腺苷有可能被用作一个再利用的抗真菌药物单独或与标准抗真菌药物的组合,并且可能是治疗真菌感染的现有疗法的快速和安全补充。
    Around 1.5 million mortality cases due to fungal infection are reported annually, posing a massive threat to global health. However, the effectiveness of current antifungal therapies in the treatment of invasive fungal infections is limited. Repurposing existing antifungal drugs is an advisable alternative approach for enhancing their effectiveness. This study evaluated the antifungal efficacy of the antiviral drug vidarabine against Candida albicans ATCC 90028. Antifungal susceptibility testing was performed by microbroth dilution assay and further processed to find the minimum fungicidal concentration. Investigation on probable mode of vidarabine action against C. albicans was assessed by using the ergosterol reduction assay, reactive oxygen species (ROS) accumulation, nuclear condensation, and apoptosis assay. Results revealed that C. albicans was susceptible to vidarabine action and exhibited minimum inhibitory concentration at 150 µg/ml. At a concentration of 300 µg/ml, vidarabine had fungicidal activity against C. albicans. 300 µg/ml vidarabine-treated C. albicans cells demonstrated 91% reduced ergosterol content. Annexin/FITC/PI assay showed that vidarabine (150 µg/ml) had increased late apoptotic cells up to 31%. As per the fractional inhibitory concentration index, vidarabine had synergistic activity with fluconazole and caspofungin against this fungus. The mechanism underlying fungicidal action of vidarabine was evaluated at the intracellular level, and probably because of increased nuclear condensation, enhanced ROS generation, and cell cycle arrest. In conclusion, this data is the first to report that vidarabine has potential to be used as a repurposed antifungal agent alone or in combination with standard antifungal drugs, and could be a quick and safe addition to existing therapies for treating fungal infections.
    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