Apilimod

APILIMOD
  • 文章类型: Journal Article
    背景:抗中性粒细胞胞浆抗体相关血管炎(AAV)是肾小球肾炎的一种快速进展形式,目前缺乏有效的治疗药物,其潜在机制尚不清楚。
    目的:本研究旨在通过结合生物信息学分析和细胞分子实验来研究AAV的新治疗方案。
    方法:该研究利用整合的生物信息学分析来鉴定具有差异表达的基因,进行富集分析,并精确定位与AAV相关的hub基因。使用ConnectivityMap和分子对接技术鉴定用于AAV的潜在治疗化合物。然后进行体外实验以检查阿吡莫德对MPO-ANCA阳性IgG诱导的内皮细胞损伤的影响和机制。
    结果:研究结果揭示了一组374个来自WGCNA差异表达基因和关键模块的常见基因,在免疫和炎症反应过程中尤其丰富。建立了蛋白质相互作用网络,导致10个枢纽基因的鉴定,包括TYROBP,PTPRC,ITGAM,KIF20A,CD86,CCL20,GAD1,LILRB2,CD8A,COL5A2来自ConnectivityMap和分子对接的分析表明,阿吡莫德可以作为基于hub基因的ANCA-GN的潜在治疗性细胞因子抑制剂。体外实验表明,阿吡莫德可以减轻紧密连接的破坏,内皮细胞通透性,LDH释放,MPO-ANCA阳性IgG诱导的内皮激活。此外,阿吡莫德治疗导致参与TLR4/NF-κB和NLRP3炎性体介导的焦亡途径的蛋白质表达显着降低。
    结论:本研究揭示了AAV的潜在发病机制,并强调了阿吡莫德通过调节TLR4/NF-kB和NLRP3炎性体介导的细胞凋亡途径减轻MPO-ANCA-IgG诱导的血管内皮细胞损伤的保护作用。这些发现表明,阿吡莫德可能有望作为AAV的治疗方法,并需要进一步调查。
    BACKGROUND: Antineutrophil cytoplasmic antibody-associated vasculitis (AAV) is a rapidly progressive form of glomerulonephritis for which effective therapeutic drugs are currently lacking, and its underlying mechanism remains unclear.
    OBJECTIVE: This study aimed to investigate new treatment options for AAV through a combination of bioinformatics analysis and cell molecular experiments.
    METHODS: The research utilized integrated bioinformatics analysis to identify genes with differential expression, conduct enrichment analysis, and pinpoint hub genes associated with AAV. Potential therapeutic compounds for AAV were identified using Connectivity Map and molecular docking techniques. In vitro experiments were then carried out to examine the impact and mechanism of apilimod on endothelial cell injury induced by MPO-ANCA-positive IgG.
    RESULTS: The findings revealed a set of 374 common genes from differentially expressed genes and key modules of WGCNA, which were notably enriched in immune and inflammatory response processes. A proteinprotein interaction network was established, leading to the identification of 10 hub genes, including TYROBP, PTPRC, ITGAM, KIF20A, CD86, CCL20, GAD1, LILRB2, CD8A, and COL5A2. Analysis from Connectivity Map and molecular docking suggested that apilimod could serve as a potential therapeutic cytokine inhibitor for ANCA-GN based on the hub genes. In vitro experiments demonstrated that apilimod could mitigate tight junction disruption, endothelial cell permeability, LDH release, and endothelial activation induced by MPO-ANCA-positive IgG. Additionally, apilimod treatment led to a significant reduction in the expression of proteins involved in the TLR4/NF-κB and NLRP3 inflammasome-mediated pyroptosis pathways.
    CONCLUSIONS: This study sheds light on the potential pathogenesis of AAV and highlights the protective role of apilimod in mitigating MPO-ANCA-IgG-induced vascular endothelial cell injury by modulating the TLR4/ NF-kB and NLRP3 inflammasome-mediated pyroptosis pathway. These findings suggest that apilimod may hold promise as a treatment for AAV and warrant further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    癌症治疗的现代医疗设备包括免疫治疗和靶向治疗,如蛋白激酶抑制剂。然而,癌症靶向药物有效动员树突状细胞(DCs)和影响免疫治疗的机制尚不清楚.这里,我们报告说,在临床相关蛋白激酶抑制剂的共有基因靶标中,在接受免疫检查点阻断(ICB)的患者中,PIKFYVE高表达对完全缓解的预测最少.在免疫细胞中,DC中PIKFYVE的高表达与ICB的不良反应相关。遗传和药理学研究表明,PIKfyve消融通过选择性改变替代/非经典NF-κB途径来增强DC功能。DC中Pikfyve的丢失和用阿吡莫德治疗,一种有效和特异性的PIKfyve抑制剂,抑制肿瘤生长,增强DC依赖性T细胞免疫,并增强了荷瘤小鼠模型中的ICB功效。此外,疫苗佐剂和阿吡莫德的组合在体内减少了肿瘤进展。因此,PIKfyve负控制DC,和PIKfyve抑制有望用于癌症免疫治疗和疫苗治疗策略。
    The modern armamentarium for cancer treatment includes immunotherapy and targeted therapy, such as protein kinase inhibitors. However, the mechanisms that allow cancer-targeting drugs to effectively mobilize dendritic cells (DCs) and affect immunotherapy are poorly understood. Here, we report that among shared gene targets of clinically relevant protein kinase inhibitors, high PIKFYVE expression was least predictive of complete response in patients who received immune checkpoint blockade (ICB). In immune cells, high PIKFYVE expression in DCs was associated with worse response to ICB. Genetic and pharmacological studies demonstrated that PIKfyve ablation enhanced DC function via selectively altering the alternate/non-canonical NF-κB pathway. Both loss of Pikfyve in DCs and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo . Thus, PIKfyve negatively controls DCs, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    胞吞途径既是细胞中分子摄取的重要途径,也是病理诱导货物的潜在切入点。细胞毒性聚集体的细胞间扩散,如帕金森病(PD)中的α-突触核蛋白(α-syn)说明了这种双重性。在这里,我们使用了人类iPSC衍生的诱导神经元模型(iNs),该模型易于通过内源性α-syn的晚期内体和溶酶体中的聚集介导死亡。由内化的预先形成的α-syn原纤维(PFF)接种。在亲本iPSC或其他非神经元细胞中未观察到这种PFF介导的死亡。使用活细胞光学显微镜观察生物传感器的读数,报告内溶酶体受伤,我们发现,iN中高达约10%的晚期内体和溶酶体表现出自发的本构穿孔,无论是否存在内在化的PFF。这个伤人,在亲本iPSC和非神经元细胞中不存在,对应于通过体积聚焦离子束扫描电子显微镜(FIB-SEM)在iN和小鼠大脑的CA1锥体神经元中直接观察到的内溶酶体子集的限制膜中的纳米孔的部分损伤,并且在iPSCs或培养物中的其他非神经元细胞或小鼠肝脏和皮肤中没有发现。我们建议iN和神经元中受损的限制膜通常是细胞溶质α-syn进入内溶酶体腔内的PFF的主要管道,启动PFF介导的α-syn聚集。重要的是,通过抑制内体磷脂酰肌醇-3-磷酸/磷脂酰肌醇5-激酶(PIKfyve激酶),使用阿吡莫德或Vuacolin-1显着减少PFF诱导的α-syn聚集,根除iN的内在内溶酶体穿孔,尽管PFF继续进入内溶酶体区室。至关重要的是,这种干预也减少了与PFF孵育相关的iN死亡。我们的结果揭示了在神经元中存在的内在穿孔的内在溶酶体,强调它们在由内化PFF诱导的毒性α-syn聚集体的形成中的关键早期参与。这一发现为采用PIKfyve激酶抑制作为潜在的治疗策略来对抗突触核蛋白病提供了基础。
    The endocytic pathway is both an essential route of molecular uptake in cells and a potential entry point for pathology-inducing cargo. The cell-to-cell spread of cytotoxic aggregates, such as those of α-synuclein (α-syn) in Parkinson\'s Disease (PD), exemplifies this duality. Here we used a human iPSC-derived induced neuronal model (iNs) prone to death mediated by aggregation in late endosomes and lysosomes of endogenous α-syn, seeded by internalized pre-formed fibrils of α-syn (PFFs). This PFF-mediated death was not observed with parental iPSCs or other non-neuronal cells. Using live-cell optical microscopy to visualize the read out of biosensors reporting endo-lysosome wounding, we discovered that up to about 10% of late endosomes and lysosomes in iNs exhibited spontaneous constitutive perforations, regardless of the presence of internalized PFFs. This wounding, absent in parental iPSCs and non-neuronal cells, corresponded to partial damage by nanopores in the limiting membranes of a subset of endolysosomes directly observed by volumetric focused ion beam scanning electron microscopy (FIB-SEM) in iNs and in CA1 pyramidal neurons from mouse brain, and not found in iPSCs or in other non-neuronal cells in culture or in mouse liver and skin. We suggest that the compromised limiting membranes in iNs and neurons in general are the primary conduit for cytosolic α-syn to access PFFs entrapped within endo-lysosomal lumens, initiating PFF-mediated α-syn aggregation. Significantly, eradicating the intrinsic endolysosomal perforations in iNs by inhibiting the endosomal Phosphatidylinositol-3-Phosphate/Phosphatidylinositol 5-Kinase (PIKfyve kinase) using Apilimod or Vacuolin-1 markedly reduced PFF-induced α-syn aggregation, despite PFFs continuing to enter the endolysosomal compartment. Crucially, this intervention also diminished iN death associated with PFF incubation. Our results reveal the surprising presence of intrinsically perforated endo-lysosomes in neurons, underscoring their crucial early involvement in the genesis of toxic α-syn aggregates induced by internalized PFFs. This discovery offers a basis for employing PIKfyve kinase inhibition as a potential therapeutic strategy to counteract synucleinopathies.
    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
    胞吞作用,或通过内体内化,是呼吸道病毒使用的主要细胞进入机制。磷酸肌醇5-激酶(PIKfyve)是合成磷脂酰肌醇(3,5)磷酸二氢盐(PtdIns(3,5)P2)的关键酶,并已通过内吞途径参与病毒运输。事实上,PIKfyve抑制剂对SARS-CoV-2和埃博拉病毒的抗病毒作用已有报道,但是关于其他呼吸道病毒的证据很少。在这项研究中,我们在体外和体内证明了PIKfyve抑制剂对流感病毒和呼吸道合胞病毒的抗病毒作用.PIKfyve抑制剂阿吡莫德甲磺酸酯(AM)和YM201636在MDCK细胞病变测定中浓度依赖性地抑制了几种流感毒株。AM也减少了病毒载量和细胞因子的释放,同时提高人鼻腔气-液界面培养的感染流感PR8的上皮细胞的完整性。在PR8感染的小鼠中,AM(2mg/mL),当鼻内治疗时,表现出病毒载量和炎症的显着减少,并抑制了由流感感染引起的体重减轻,效果类似于口服奥司他韦(10mg/kg)。此外,AM在体外和体内小鼠中在RSVA2感染的人鼻上皮中表现出抗病毒作用,效果与利巴韦林相当。AM还在体外显示出对人类鼻病毒和季节性冠状病毒的抗病毒作用。因此,PIKfyve被发现与流感和RSV感染有关,和PIKfyve抑制剂是一种有前途的分子,用于抗呼吸道病毒的泛病毒方法。
    Endocytosis, or internalization through endosomes, is a major cell entry mechanism used by respiratory viruses. Phosphoinositide 5-kinase (PIKfyve) is a critical enzyme for the synthesis of phosphatidylinositol (3, 5)biphosphate (PtdIns (3, 5)P2) and has been implicated in virus trafficking via the endocytic pathway. In fact, antiviral effects of PIKfyve inhibitors against SARS-CoV-2 and Ebola have been reported, but there is little evidence regarding other respiratory viruses. In this study, we demonstrated the antiviral effects of PIKfyve inhibitors on influenza virus and respiratory syncytial virus in vitro and in vivo. PIKfyve inhibitors Apilimod mesylate (AM) and YM201636 concentration-dependently inhibited several influenza strains in an MDCK cell-cytopathic assay. AM also reduced the viral load and cytokine release, while improving the cell integrity of human nasal air-liquid interface cultured epithelium infected with influenza PR8. In PR8-infected mice, AM (2 mg/mL), when intranasally treated, exhibited a significant reduction of viral load and inflammation and inhibited weight loss caused by influenza infection, with effects being similar to oral oseltamivir (10 mg/kg). In addition, AM demonstrated antiviral effects in RSV A2-infected human nasal epithelium in vitro and mouse in vivo, with an equivalent effect to that of ribavirin. AM also showed antiviral effects against human rhinovirus and seasonal coronavirus in vitro. Thus, PIKfyve is found to be involved in influenza and RSV infection, and PIKfyve inhibitor is a promising molecule for a pan-viral approach against respiratory viruses.
    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
    NLRP3是一种重要的先天性免疫传感器,可响应各种信号并形成炎症复合体,导致IL-1β分泌和焦亡。溶酶体损伤与响应晶体或颗粒的NLRP3炎性体激活有关,但机制尚不清楚。我们开发了小分子文库筛选,发现阿吡莫德,溶酶体干扰物,是选择性和有效的NLRP3激动剂。阿吡莫德促进NLRP3炎性体激活,IL-1β分泌,和焦亡。机械上,而阿吡莫德对NLRP3的激活独立于钾外排和直接结合,阿吡莫德引发线粒体损伤和溶酶体功能障碍。此外,我们发现阿吡莫德在溶酶体中诱导TRPML1依赖性钙通量,导致线粒体损伤和NLRP3炎性体激活。因此,我们的结果揭示了阿吡莫德的促炎性体活性和钙依赖性溶酶体介导的NLRP3炎性体激活的机制。
    NLRP3 is an important innate immune sensor that responses to various signals and forms the inflammasome complex, leading to IL-1β secretion and pyroptosis. Lysosomal damage has been implicated in NLRP3 inflammasome activation in response to crystals or particulates, but the mechanism remains unclear. We developed the small molecule library screening and found that apilimod, a lysosomal disruptor, is a selective and potent NLRP3 agonist. Apilimod promotes the NLRP3 inflammasome activation, IL-1β secretion, and pyroptosis. Mechanismically, while the activation of NLRP3 by apilimod is independent of potassium efflux and directly binding, apilimod triggers mitochondrial damage and lysosomal dysfunction. Furthermore, we found that apilimod induces TRPML1-dependent calcium flux in lysosomes, leading to mitochondrial damage and the NLRP3 inflammasome activation. Thus, our results revealed the pro-inflammasome activity of apilimod and the mechanism of calcium-dependent lysosome-mediated NLRP3 inflammasome activation.
    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
    2019冠状病毒病(COVID-19)是由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)引起的疾病。除了呼吸道疾病,COVID-19患者表现出持续数周至数月的神经症状(长COVID)。尚不清楚这些神经系统表现是否是由于脑细胞感染所致。我们发现一小部分人诱导多能干细胞(iPSC)衍生的神经元,但不是星形胶质细胞,自然易患SARS-CoV-2。基于阻断抗体的抑制作用,感染似乎依赖于受体血管紧张素转换酶2(ACE2),尽管其在神经元中的表达水平非常低。细胞质中双链RNA的存在(病毒复制的标志),病毒晚期基因的丰富合成位于整个感染细胞,并且在感染的前48小时内培养基中病毒RNA水平的增加(病毒释放)表明感染是生产性的。SARS-CoV-2的生产性进入需要病毒和细胞膜的融合,这导致病毒基因组递送到靶细胞的细胞质中。融合是由病毒表面刺突蛋白的蛋白水解裂解引发的,它可以发生在质膜或从内体或溶酶体。我们发现SARS-CoV-2感染人类神经元对nafamostat和camostat不敏感,抑制细胞丝氨酸蛋白酶,包括跨膜丝氨酸蛋白酶2(TMPRSS2)。组织蛋白酶L的抑制也没有显著阻断感染。相比之下,神经元感染被阿吡莫德阻断,磷脂酰肌醇5激酶(PIK5K)的抑制剂,调节内体成熟的早期到晚期。重要性COVID-19是一种由冠状病毒SARS-CoV-2引起的疾病。数百万患者表现出神经症状,包括头痛,记忆受损,癫痫发作,和脑病,以及解剖异常,比如大脑形态的变化。SARS-CoV-2感染人脑已被证明,但目前尚不清楚观察到的神经症状是否与直接的脑部感染有关。病毒进入神经元的机制也没有被表征。这里,我们使用人类iPSC来源的神经细胞模型研究了SARS-CoV-2感染,发现一小部分皮质样神经元天然易感感染.生产性感染是ACE2依赖性和TMPRSS2依赖性。我们还发现该病毒使用晚期内体和溶酶体途径进入细胞,并且感染可以被阿吡莫德阻断,细胞PIK5K的抑制剂。
    2019 coronavirus disease (COVID-19) is a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to respiratory illness, COVID-19 patients exhibit neurological symptoms lasting from weeks to months (long COVID). It is unclear whether these neurological manifestations are due to an infection of brain cells. We found that a small fraction of human induced pluripotent stem cell (iPSC)-derived neurons, but not astrocytes, were naturally susceptible to SARS-CoV-2. Based on the inhibitory effect of blocking antibodies, the infection seemed to depend on the receptor angiotensin-converting enzyme 2 (ACE2), despite very low levels of its expression in neurons. The presence of double-stranded RNA in the cytoplasm (the hallmark of viral replication), abundant synthesis of viral late genes localized throughout infected cells, and an increase in the level of viral RNA in the culture medium (viral release) within the first 48 h of infection suggested that the infection was productive. Productive entry of SARS-CoV-2 requires the fusion of the viral and cellular membranes, which results in the delivery of the viral genome into the cytoplasm of the target cell. The fusion is triggered by proteolytic cleavage of the viral surface spike protein, which can occur at the plasma membrane or from endosomes or lysosomes. We found that SARS-CoV-2 infection of human neurons was insensitive to nafamostat and camostat, which inhibit cellular serine proteases, including transmembrane serine protease 2 (TMPRSS2). Inhibition of cathepsin L also did not significantly block infection. In contrast, the neuronal infection was blocked by apilimod, an inhibitor of phosphatidyl-inositol 5 kinase (PIK5K), which regulates early to late endosome maturation. IMPORTANCE COVID-19 is a disease caused by the coronavirus SARS-CoV-2. Millions of patients display neurological symptoms, including headache, impairment of memory, seizures, and encephalopathy, as well as anatomical abnormalities, such as changes in brain morphology. SARS-CoV-2 infection of the human brain has been documented, but it is unclear whether the observed neurological symptoms are linked to direct brain infection. The mechanism of virus entry into neurons has also not been characterized. Here, we investigated SARS-CoV-2 infection by using a human iPSC-derived neural cell model and found that a small fraction of cortical-like neurons was naturally susceptible to infection. The productive infection was ACE2 dependent and TMPRSS2 independent. We also found that the virus used the late endosomal and lysosomal pathway for cell entry and that the infection could be blocked by apilimod, an inhibitor of cellular PIK5K.
    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
    溶酶体膜含有协调溶酶体功能和动力学的多种磷酸肌醇(PtdIns)脂质。溶酶体上的PtdIns库受到多种PtdIns激酶和磷酸酶的作用的严格调节;然而,PtdIns在溶酶体功能和动力学中的具体作用目前尚不清楚,需要进一步研究.以前表明,PIKfyve,从PtdIns(3)P合成PtdIns(3,5)P2的脂质激酶,控制溶酶体“聚变裂变”循环动力学,自噬体周转,和内吞货物运输。此外,INPP4B,水解PtdIns(3,4)P2形成PtdIns(3)P的PtdIns4-磷酸酶,正在成为一种在溶酶体生物发生和其他溶酶体功能中起作用的癌症相关蛋白。这里,我们研究了在Inpp4b缺陷小鼠胚胎成纤维细胞中破坏PIKfyve功能的后果.通过共聚焦荧光成像,我们观察到大量扩大的溶酶体的形成,伴随着内吞贩运的加剧减少,破坏溶酶体融合-裂变动力学,和抑制自噬。最后,3H-肌醇标记的PtdIns和PtdIns免疫荧光染色的HPLC闪烁定量,我们观察到,由于磷脂酰肌醇3-激酶催化亚基VPS34酶活性的过度激活,溶酶体PtdIns(3)P水平在Inpp4b缺陷型细胞中显著升高.总之,我们的研究确定了一个新的信号轴,维持正常的溶酶体稳态和动力学,其中包括Inpp4b的催化功能,PIKfyve,VPS34
    Lysosome membranes contain diverse phosphoinositide (PtdIns) lipids that coordinate lysosome function and dynamics. The PtdIns repertoire on lysosomes is tightly regulated by the actions of diverse PtdIns kinases and phosphatases; however, specific roles for PtdIns in lysosomal functions and dynamics are currently unclear and require further investigation. It was previously shown that PIKfyve, a lipid kinase that synthesizes PtdIns(3,5)P2 from PtdIns(3)P, controls lysosome \"fusion-fission\" cycle dynamics, autophagosome turnover, and endocytic cargo delivery. Furthermore, INPP4B, a PtdIns 4-phosphatase that hydrolyzes PtdIns(3,4)P2 to form PtdIns(3)P, is emerging as a cancer-associated protein with roles in lysosomal biogenesis and other lysosomal functions. Here, we investigated the consequences of disrupting PIKfyve function in Inpp4b-deficient mouse embryonic fibroblasts. Through confocal fluorescence imaging, we observed the formation of massively enlarged lysosomes, accompanied by exacerbated reduction of endocytic trafficking, disrupted lysosome fusion-fission dynamics, and inhibition of autophagy. Finally, HPLC scintillation quantification of 3H-myo-inositol labeled PtdIns and PtdIns immunofluorescence staining, we observed that lysosomal PtdIns(3)P levels were significantly elevated in Inpp4b-deficient cells due to the hyperactivation of phosphatidylinositol 3-kinase catalytic subunit VPS34 enzymatic activity. In conclusion, our study identifies a novel signaling axis that maintains normal lysosomal homeostasis and dynamics, which includes the catalytic functions of Inpp4b, PIKfyve, and VPS34.
    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
    原理:TGFβ信号通路控制组织纤维化重塑,许多导致器官损伤和衰竭的疾病的标志。在这项研究中,我们讨论阿吡莫德的作用,脂质激酶PIKfyve的药理学抑制剂,在心脏病理性纤维化重塑和TGFβ信号通路的调控中。方法:阿吡莫德治疗对心肌纤维化的影响,在压力超负荷诱导的心力衰竭小鼠模型中体内评估肥大和心功能.用Apilimod处理的原代心脏成纤维细胞和HeLa细胞以及小鼠胚胎成纤维细胞中PIKfyve的基因突变被用作细胞模型。结果:体内给药时,阿吡莫德减少心肌间质纤维化发展并预防左心室功能障碍。体外,阿吡莫德控制原代鼠心脏成纤维细胞的TGFβ依赖性活化。机械上,Apilimod和PIKfyve的基因突变均可诱导细胞内囊泡中的TGFβ受体阻断,负调节其下游信号通路并最终抑制TGFβ反应。结论:总之,我们的发现提出了PIKfyve在控制心肌纤维化重塑和TGFβ信号通路中的新功能,因此,为使用阿吡莫德治疗预防不良纤维化重塑的新治疗观点开辟了道路。
    Rationale: TGFβ signaling pathway controls tissue fibrotic remodeling, a hallmark in many diseases leading to organ injury and failure. In this study, we address the role of Apilimod, a pharmacological inhibitor of the lipid kinase PIKfyve, in the regulation of cardiac pathological fibrotic remodeling and TGFβ signaling pathway. Methods: The effects of Apilimod treatment on myocardial fibrosis, hypertrophy and cardiac function were assessed in vivo in a mouse model of pressure overload-induced heart failure. Primary cardiac fibroblasts and HeLa cells treated with Apilimod as well as genetic mutation of PIKfyve in mouse embryonic fibroblasts were used as cell models. Results: When administered in vivo, Apilimod reduced myocardial interstitial fibrosis development and prevented left ventricular dysfunction. In vitro, Apilimod controlled TGFβ-dependent activation of primary murine cardiac fibroblasts. Mechanistically, both Apilimod and genetic mutation of PIKfyve induced TGFβ receptor blockade in intracellular vesicles, negatively modulating its downstream signaling pathway and ultimately dampening TGFβ response. Conclusions: Altogether, our findings propose a novel function for PIKfyve in the control of myocardial fibrotic remodeling and the TGFβ signaling pathway, therefore opening the way to new therapeutic perspectives to prevent adverse fibrotic remodeling using Apilimod treatment.
    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
    埃博拉病毒(EBOV)的爆发与高发病率和死亡率有关。最近在EBOV疾病(EVD)的管理方面已经达到了里程碑,并获得了EBOV疫苗和两种单克隆抗体疗法的许可。然而,对于其他致病丝状病毒,既没有疫苗也没有治疗方法。为了准备这种爆发,为了更容易和更具成本效益地管理EVD,我们寻求一种含有口服和室温稳定的药物的鸡尾酒,对多种丝状病毒具有很强的活性。我们先前表明(贝普地尔舍曲林)和(舍曲林托瑞米芬)协同抑制细胞培养物中的EBOV。这里,我们描述了在EVD小鼠模型中测试这些组合的步骤.我们确定了一种适合口服给药的载体,如此配制的药物与DMSO中的制剂一样具有相同的抗EBOV活性,它们在溶液中储存后保持活性长达七天。药代动力学(PK)研究表明,口服递送载体中的药物在测试的最高剂量下在小鼠中是良好耐受的。数据总体上支持这些组合在EVD小鼠模型中测试协同作用的进展。此外,基于人类口服PK的数学模型预测,这些组合在人类中的活性比其成分单一药物更高。
    Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The PIKfyve inhibitor apilimod is currently undergoing clinical trials for treatment of COVID-19. However, although apilimod might prevent viral invasion by inhibiting host cell proteases, the same proteases are critical for antigen presentation leading to T cell activation and there is good evidence from both in vitro studies and the clinic that apilimod blocks antiviral immune responses. We therefore warn that the immunosuppression observed in many COVID-19 patients might be aggravated by apilimod.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号