PAK1

PAK1
  • 文章类型: Journal Article
    与痉挛相关的反射亢进是一种普遍的神经系统疾病,其特征是对刺激的过度和过度的反射反应。反射亢进可由多种疾病引起,包括多发性硬化症,中风和脊髓损伤(SCI)。尽管我们以前已经确定了脊髓反射亢进与SCI诱导的痉挛有关的RAC1-PAK1通路的贡献,可行的药物靶标尚未得到验证.为了评估靶向PAK1减弱H反射过度兴奋的效用,我们给Romidepsin,临床上可用的PAK1抑制剂,在Thy1-YFP报告小鼠中。我们进行了纵向肌电图研究,研究设计使我们能够评估病理性H反射变化和药物干预效果随着时间的推移,挫伤SCI之前和之后。不出所料,我们的结果表明,与基线相比,SCI后1个月,速率依赖性抑郁症-反射亢进和痉挛的迹象-显着丧失。未受伤的控制(或受伤前)。与对照组相比,在SCI动物的药物干预前后,Romidepsin治疗减少了反射亢进的迹象。神经解剖学研究进一步证实了药物反应,因为罗米地辛治疗也减少了SCI诱导的树突状脊柱发育不全在α-运动神经元上的存在。一起来看,我们的发现扩展了先前的工作,证明了在SCI诱导的痉挛中靶向PAK1活性的效用,并支持romidepsin作为控制痉挛的有效工具的新颖用途.关键点:PAK1通过促进树突状脊柱发育不全,在脊髓损伤(SCI)引起的痉挛中发挥作用。在这项研究中,我们探讨了在SCI小鼠模型中抑制PAK1减少痉挛和树突状脊柱发育不全的临床前效用.Romidepsin是2009年在美国批准用于治疗皮肤T细胞淋巴瘤的PAK1抑制剂。在这里,我们表明SCI后的罗米地辛治疗降低了SCI诱导的H反射过度兴奋性和异常的α运动神经元脊柱形态。这项研究提供了令人信服的证据,表明罗米地辛可能是减轻SCI引起的痉挛的有希望的治疗方法。
    Hyperreflexia associated with spasticity is a prevalent neurological condition characterized by excessive and exaggerated reflex responses to stimuli. Hyperreflexia can be caused by several diseases including multiple sclerosis, stroke and spinal cord injury (SCI). Although we have previously identified the contribution of the RAC1-PAK1 pathway underlying spinal hyperreflexia with SCI-induced spasticity, a feasible druggable target has not been validated. To assess the utility of targeting PAK1 to attenuate H-reflex hyperexcitability, we administered Romidepsin, a clinically available PAK1 inhibitor, in Thy1-YFP reporter mice. We performed longitudinal EMG studies with a study design that allowed us to assess pathological H-reflex changes and drug intervention effects over time, before and after contusive SCI. As expected, our results show a significant loss of rate-dependent depression - an indication of hyperreflexia and spasticity - 1 month following SCI as compared with baseline, uninjured controls (or before injury). Romidepsin treatment reduced signs of hyperreflexia in comparison with control cohorts and in pre- and post-drug intervention in SCI animals. Neuroanatomical study further confirmed drug response, as romidepsin treatment also reduced the presence of SCI-induced dendritic spine dysgenesis on α-motor neurons. Taken together, our findings extend previous work demonstrating the utility of targeting PAK1 activity in SCI-induced spasticity and support the novel use of romidepsin as an effective tool for managing spasticity. KEY POINTS: PAK1 plays a role in contributing to the development of spinal cord injury (SCI)-induced spasticity by contributing to dendritic spine dysgenesis. In this study, we explored the preclinical utility of inhibiting PAK1 to reduce spasticity and dendritic spine dysgenesis in an SCI mouse model. Romidepsin is a PAK1 inhibitor approved in the US in 2009 for the treatment of cutaneous T-cell lymphoma. Here we show that romidepsin treatment after SCI reduced SCI-induced H-reflex hyperexcitability and abnormal α-motor neuron spine morphology. This study provides compelling evidence that romidepsin may be a promising therapeutic approach for attenuating SCI-induced spasticity.
    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
    三阴性乳腺癌是临床上最恶性的亚型之一,迫切需要寻找新的治疗方法。p21激活的激酶I(PAK1)被认为是TNBC的有吸引力的治疗靶标。在这项研究中,我们通过将VHL或CRBN连接酶配体与烷基链或PEG链连接的PAK1抑制剂缀合,设计并合成了一系列新型PROTACPAK1降解剂。最有前途的化合物,19s,可以在低至0.1μM的浓度下显著降解PAK1蛋白,并在MDA-MB-231细胞中实现了有效的抗增殖活性,IC50值为1.27μM。此外,19s在体外表现出有效的抗迁移活性,并在体内诱导快速的肿瘤消退。总的来说,这些发现证明19s是一种有效的新型PAK1降解剂,具有治疗TNBC的潜力。
    Triple-negative breast cancer is one of the most malignant subtypes in clinical practice, and it is urgent to find new therapies. The p21-activated kinase I (PAK1) has been considered to be an attractive therapeutic target for TNBC. In this study, we designed and synthesized a series of novel PROTAC PAK1 degraders by conjugating VHL or CRBN ligase ligands to PAK1 inhibitors which are connected by alkyl chains or PEG chains. The most promising compound, 19s, can significantly degrade PAK1 protein at concentrations as low as 0.1 μM, and achieves potent anti-proliferative activity with an IC50 value of 1.27 μM in MDA-MB-231 cells. Additionally, 19s exhibits potent anti-migration activity in vitro and induces rapid tumor regression in vivo. Collectively, these findings document that 19s is a potent and novel PAK1 degrader with promising potential for TNBC 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
    背景:越来越多的研究证明了环状RNA(circularRNAs,circRNAs)与各种疾病的病理过程以及它们参与多种癌症的发作和进展的关联。然而,circRNAs在胃癌自噬调控中的功能作用和潜在机制尚未完全阐明。
    方法:我们使用透射电子显微镜和mRFP-GFP-LC3双荧光自噬指示剂来研究自噬调节。细胞计数试剂盒-8测定,集落形成试验,5-乙炔基-2'-脱氧尿苷掺入测定,Transwell分析,并进行蛋白质印迹分析以确认circPTPN22对GC进展的影响。双重荧光素酶报告基因试验验证了circPTPN22和miR-6788-5p之间的结合,以及miR-6788-5p和p21激活的激酶-1(PAK1)。功能拯救实验评估circPTPN22是否通过竞争性结合miR-6788-5p调节PAK1表达,影响GC细胞的自噬和其他生物学过程。我们使用裸鼠异种移植模型研究了circPTPN22对体内GC肿瘤的影响。生物信息学工具预测了circPTPN22的上游调节转录因子和结合蛋白,而染色质免疫沉淀和核糖核蛋白免疫沉淀测定证实了结合状态。
    结果:在GC中上调circPTPN22已被证明可以抑制自噬并促进细胞增殖,迁移,和入侵。机械上,circPTPN22直接结合miR-6788-5p,随后调节PAK1的表达,从而激活蛋白激酶B(Akt)和细胞外信号调节激酶(Erk)的磷酸化。这种调节最终影响GC细胞中的自噬水平。此外,runt相关转录因子1(RUNX1)负调控circPTPN22表达,而RNA结合蛋白如FUS(融合在肉瘤中)和ELAVL1(重组ELAV样蛋白1)正调节其表达。抑制自噬途径可增加FUS表达,进一步上调GC细胞中的circPTPN22,从而加剧GC的进展。
    结论:在转录因子RUNX1和RNA结合蛋白FUS和ELAVL1的调控下,circPTPN22通过miR-6788-5p/PAK1轴激活Akt和Erk的磷酸化,从而调节GC细胞中的自噬。抑制自噬增加FUS,进而在PTPN22周围上调,形成正反馈回路,最终加速GC的进展。
    BACKGROUND: An increasing number of studies have demonstrated the association of circular RNAs (circRNAs) with the pathological processes of various diseases and their involvement in the onset and progression of multiple cancers. Nevertheless, the functional roles and underlying mechanisms of circRNAs in the autophagy regulation of gastric cancer (GC) have not been fully elucidated.
    METHODS: We used transmission electron microscopy and the mRFP-GFP-LC3 dual fluorescent autophagy indicator to investigate autophagy regulation. The cell counting kit-8 assay, colony formation assay, 5-ethynyl-2\'-deoxyuridine incorporation assay, Transwell assay, and Western blot assay were conducted to confirm circPTPN22\'s influence on GC progression. Dual luciferase reporter assays validated the binding between circPTPN22 and miR-6788-5p, as well as miR-6788-5p and p21-activated kinase-1 (PAK1). Functional rescue experiments assessed whether circPTPN22 modulates PAK1 expression by competitively binding miR-6788-5p, affecting autophagy and other biological processes in GC cells. We investigated the impact of circPTPN22 on in vivo GC tumors using a nude mouse xenograft model. Bioinformatics tools predicted upstream regulatory transcription factors and binding proteins of circPTPN22, while chromatin immunoprecipitation and ribonucleoprotein immunoprecipitation assays confirmed the binding status.
    RESULTS: Upregulation of circPTPN22 in GC has been shown to inhibit autophagy and promote cell proliferation, migration, and invasion. Mechanistically, circPTPN22 directly binds to miR-6788-5p, subsequently regulating the expression of PAK1, which activates protein kinase B (Akt) and extracellular signal-regulated kinase (Erk) phosphorylation. This modulation ultimately affects autophagy levels in GC cells. Additionally, runt-related transcription factor 1 (RUNX1) negatively regulates circPTPN22 expression, while RNA-binding proteins such as FUS (fused in sarcoma) and ELAVL1 (recombinant ELAV-like protein 1) positively regulate its expression. Inhibition of the autophagy pathway can increase FUS expression, further upregulating circPTPN22 in GC cells, thereby exacerbating the progression of GC.
    CONCLUSIONS: Under the regulation of the transcription factor RUNX1 and RNA-binding proteins FUS and ELAVL1, circPTPN22 activates the phosphorylation of Akt and Erk through the miR-6788-5p/PAK1 axis, thereby modulating autophagy in GC cells. Inhibition of autophagy increases FUS, which in turn upregulates circPTPN22, forming a positive feedback loop that ultimately accelerates the progression of GC.
    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
    在中枢神经系统中,少突胶质细胞(OLs)形成髓鞘依赖于肌动蛋白细胞骨架聚合向解聚的转变。触发这种开关的分子机制尚未阐明。这里,我们确定P21激活的激酶1(PAK1)是OLs中肌动蛋白解聚的主要调节因子。我们的结果表明,PAK1以激酶抑制的形式在OLs中积累,触发肌动蛋白拆卸和,因此,髓鞘膜扩张.值得注意的是,PAK1结合配偶体的蛋白质组学分析能够鉴定NF2/Merlin作为其内源性抑制剂。我们的发现表明,在OLs中Nf2敲低导致PAK1激活,肌动蛋白聚合,和髓磷脂膜扩张的减少。通过用PAK1抑制剂治疗来挽救这种效应。我们还提供证据表明,少突胶质细胞中特定的Pak1功能丧失会刺激体内髓鞘的增厚。总的来说,我们的数据表明,PAK1和NF2/Merlin对OL的肌动蛋白细胞骨架的拮抗作用对于正常的髓磷脂形成至关重要。这些发现在脱髓鞘疾病和神经发育障碍中具有广泛的机制和治疗意义。
    In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.
    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
    骨质疏松症(OP)发病机制的复杂编排仍然难以捉摸。越来越多的证据表明,血管生成驱动的成骨作用是维持骨稳态的关键基础。本研究旨在探讨内皮血小板衍生生长因子受体-β(PDGFR-β)通过促进H型血管形成来减轻骨丢失的潜力。我们的发现表明,在从患有OP的个体获得的样本中,内皮PDGFR-β的表达水平降低。以及卵巢切除小鼠。内皮细胞中PDGFR-β的消耗改善了小鼠血管生成介导的骨形成。内皮PDGFR-β对H型血管的调节影响是通过PDGFRβ-P21激活的激酶1-Notch1细胞内结构域信号级联介导的。特别是,PDGFR-β的内皮特异性增强促进了OP中H型血管及其相关骨的形成。因此,在不久的将来,靶向内皮PDGFR-β的策略成为治疗OP的一种有前景的治疗方法.
    The intricate orchestration of osteoporosis (OP) pathogenesis remains elusive. Mounting evidence suggests that angiogenesis-driven osteogenesis serves as a crucial foundation for maintaining bone homeostasis. This study aimed to explore the potential of the endothelial platelet-derived growth factor receptor-β (PDGFR-β) in mitigating bone loss through its facilitation of H-type vessel formation. Our findings demonstrate that the expression level of endothelial PDGFR-β is reduced in samples obtained from individuals suffering from OP, as well as in ovariectomy mice. Depletion of PDGFR-β in endothelial cells ameliorates angiogenesis-mediated bone formation in mice. The regulatory influence of endothelial PDGFR-β on H-type vessels is mediated through the PDGFRβ-P21-activated kinase 1-Notch1 intracellular domain signaling cascade. In particular, the endothelium-specific enhancement of PDGFR-β facilitates H-type vessels and their associated bone formation in OP. Hence, the strategic targeting of endothelial PDGFR-β emerges as a promising therapeutic approach for the management of OP in the near future.
    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
    PAK1(RAC/CDC42激活激酶1)和TOR(雷帕霉素靶标)都是主要的致癌/老化激酶。然而,它们在我们的免疫系统中起着相反的作用,即免疫系统被PAK1抑制,而它需要TOR。因此,PAK1-阻断剂,对癌症的治疗更有效,比TOR-阻滞剂。自2015年以来,我们发现PDGF诱导的黑色素生成依赖于“PAK1”我们能够筛选出一系列PAK1阻断剂作为黑色素生成抑制剂,最终可以促进寿命.有趣的是,雷帕霉素,第一种TOR抑制剂,促进黑色素生成,清楚地表明TOR抑制黑色素生成。然而,一种名为TORin-1的新TOR抑制剂不再抑制免疫系统,并阻断细胞培养中的黑色素生成。这些观察强烈表明TORin-1作为PAK1受体阻滞剂,而不是TOR阻滞剂,在体内。因此,细胞培养中的黑素生成很可能使我们能够区分PAK1阻断剂和TORblockers.
    Both PAK1 (RAC/CDC42-activating kinase 1) and TOR (Target of Rapamycin) are among the major oncogenic/ageing kinases. However, they play the opposite role in our immune system, namely immune system is suppressed by PAK1, while it requires TOR. Thus, PAK1-blockers, would be more effective for therapy of cancers, than TOR-blockers. Since 2015 when we discovered genetically that PDGF-induced melanogenesis depends on \"PAK1\", we are able to screening a series of PAK1-blockers as melanogenesis-inhibitors which could eventually promote longevity. Interestingly, rapamycin, the first TOR-inhibitor, promotes melanogenesis, clearly indicating that TOR suppresses melanogenesis. However, a new TOR-inhibitor called TORin-1 no longer suppresses immune system, and blocks melanogenesis in cell culture. These observations strongly indicate that TORin-1 acts as PAK1-blockers, instead of TOR-blockers, in vivo. Thus, it is most likely that melanogenesis in cell culture could enable us to discriminate PAK1-blockers from TORblockers.
    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
    脓毒症是一种以器官功能障碍为特征的严重综合征,由于对感染的反应引起的全身失衡。PAK1在各种疾病中起关键作用。本研究旨在探讨PAK1在脓毒症炎症中的作用机制。进行生物信息学分析以评估PAK1,蜗牛,和CXCL2在脓毒症患者全血中的表达以及PAK1富集的通路。为了模拟脓毒症模型,用脂多糖刺激THP-1细胞。使用qRT-PCR评估基因表达,而使用CCK-8测定评估细胞活力。用流式细胞术检测细胞凋亡。使用酶联免疫吸附测定(ELISA)分析不同处理后细胞中炎性因子的表达。进行双荧光素酶和染色质免疫沉淀测定以验证PAK1与蜗牛之间的结合关系。建立小鼠盲肠结扎穿刺术模型,用苏木精、伊红染色和ELISA检测肺组织炎症细胞浸润水平及相关保护因子的表达,肝脏,和肾脏组织。结果表明PAK1,蜗牛,和脓毒症患者全血中的CXCL2,PAK1富集在趋化因子相关途径中。敲除PAK1可显著促进LPS刺激的THP-1细胞凋亡,抑制炎症因子的表达。PAK1上调蜗牛的表达,进而促进CXCL2的表达。因此,PAK1通过snail/CXCL2通路介导脓毒症诱导的炎症反应。总之,PAK1通过蜗牛/CXCL2轴促进脓毒症诱导的炎症反应,从而为脓毒症的管理提供了潜在的治疗靶标。
    Sepsis is a severe syndrome characterized by organ dysfunction, resulting from a systemic imbalance in response to infection. PAK1 plays a critical role in various diseases. The present study aimed to explore and delineate the mechanism of PAK1 in inflammation induced by sepsis. Bioinformatics analysis was performed to assess PAK1, snail, and CXCL2 expression in the whole blood of septic patients and the pathways enriched with PAK1. To simulate the sepsis model, THP-1 cells were stimulated with lipopolysaccharide. Gene expression was evaluated using qRT-PCR, while cell viability was assessed using CCK-8 assay. Cell apoptosis was tested with flow cytometry. Expression of inflammatory factors in cells following different treatments was analyzed using the enzyme linked immunosorbent assay (ELISA). Dual-luciferase and chromatin immunoprecipitation assays were conducted to verify the binding relationship between PAK1 and the snail. Mouse models of cecal ligation and puncture were established, and hematoxylin and eosin staining and ELISA were employed to detect the infiltration levels of inflammatory cells and the expression of related protective factors in lung, liver, and kidney tissues. The results demonstrated upregulation of PAK1, snail, and CXCL2 in the whole blood of septic patients, with PAK1 being enriched in the chemokine-related pathway. Knockdown of PAK1 significantly promoted the apoptosis of LPS-stimulated THP-1 cells and inhibited the expression of inflammatory factors. PAK1 upregulated the expression of the snail, which in turn promoted the expression of CXCL2. Thus, PAK1 mediated the sepsis-induced inflammatory response through the snail/CXCL2 pathway. In conclusion, PAK1 played a role in promoting inflammation induced by sepsis through the snail/CXCL2 axis, thereby providing a potential therapeutic target for the management of sepsis.
    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
    肾足细胞的功能与Rho小GTP酶调节的肌动蛋白细胞骨架密切相关。肌动蛋白驱动的细胞粘附和过程的丧失与足细胞功能障碍有关,蛋白尿,和肾脏疾病。FilGAP,Rho小GTPaseRac1的GTPase激活蛋白在肾足细胞中大量表达,它的基因与局灶性节段性肾小球硬化家族的疾病有关。在这项研究中,我们已经研究了FilGAP在体外足细胞中的作用。FilGAP在培养的足细胞中的消耗导致肌动蛋白应力纤维的损失和Rac1活性的增加。相反,FilGAP的强制表达增加了应力纤维的形成,而Rac1的激活显着减少了其形成。FilGAP位于局灶性粘连(FA),与应力纤维密切相关的基于整合素的蛋白质复合物,介导细胞-细胞外基质(ECM)粘附,FilGAP耗竭减少了FA形成和对ECM的附着受损。此外,在独特的足细胞细胞培养物中,能够诱导形成高度组织的过程,包括主要过程和足过程样突起,FilGAP耗尽或Rac1活化减少了这些过程的形成。通过抑制Rac1或P21激活的激酶1(PAK1),可以挽救FilGAP耗尽的足细胞中FAs的减少和过程形成,Rac1的下游效应物和PAK1激活抑制了它们的形成。因此,FilGAP通过抑制Rac1/PAK1信号传导有助于细胞-ECM粘附和足细胞的过程形成。
    The function of kidney podocytes is closely associated with actin cytoskeleton regulated by Rho small GTPases. Loss of actin-driven cell adhesions and processes is connected to podocyte dysfunction, proteinuria, and kidney diseases. FilGAP, a GTPase-activating protein for Rho small GTPase Rac1, is abundantly expressed in kidney podocytes, and its gene is linked to diseases in a family with focal segmental glomerulosclerosis. In this study, we have studied the role of FilGAP in podocytes in vitro. Depletion of FilGAP in cultured podocytes induced loss of actin stress fibers and increased Rac1 activity. Conversely, forced expression of FilGAP increased stress fiber formation whereas Rac1 activation significantly reduced its formation. FilGAP localizes at the focal adhesion (FA), an integrin-based protein complex closely associated with stress fibers, that mediates cell-extracellular matrix (ECM) adhesion, and FilGAP depletion decreased FA formation and impaired attachment to the ECM. Moreover, in unique podocyte cell cultures capable of inducing the formation of highly organized processes including major processes and foot process-like projections, FilGAP depletion or Rac1 activation decreased the formation of these processes. The reduction of FAs and process formations in FilGAP-depleted podocyte cells was rescued by inhibition of Rac1 or P21-activated kinase 1 (PAK1), a downstream effector of Rac1, and PAK1 activation inhibited their formations. Thus, FilGAP contributes to both cell-ECM adhesion and process formation of podocytes by suppressing Rac1/PAK1 signaling.
    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
    VAV2是RHOGTP酶的激活剂,可促进和维持正常角质形成细胞和口腔鳞状细胞癌(OSCC)细胞的再生增殖样状态。这里,我们证明VAV2还调节这些细胞中的核糖体生物发生,与人乳头瘤病毒阴性(HPV-)OSCC患者预后不良相关的项目。机械上,VAV2使用包含RAC1和RHOAGTP酶的保守途径以催化依赖性方式调节该过程,PAK和ROCK家族激酶,以及c-MYC和YAP/TAZ转录因子。该途径直接促进RNA聚合酶I活性和47S前rRNA前体的合成。核糖体生物发生因子的上调和YAP/TAZ依赖性未分化细胞状态的获得进一步巩固了该过程。最后,我们显示RNA聚合酶I是角质形成细胞和OSCC患者来源的细胞的治疗性致命弱点,具有高VAV2催化活性。总的来说,这些发现强调了在OSCC的肿瘤前期和晚期进展阶段调节VAV2和核糖体生物发生途径的治疗潜力.
    VAV2 is an activator of RHO GTPases that promotes and maintains regenerative proliferation-like states in normal keratinocytes and oral squamous cell carcinoma (OSCC) cells. Here, we demonstrate that VAV2 also regulates ribosome biogenesis in those cells, a program associated with poor prognosis of human papilloma virus-negative (HPV-) OSCC patients. Mechanistically, VAV2 regulates this process in a catalysis-dependent manner using a conserved pathway comprising the RAC1 and RHOA GTPases, the PAK and ROCK family kinases, and the c-MYC and YAP/TAZ transcription factors. This pathway directly promotes RNA polymerase I activity and synthesis of 47S pre-rRNA precursors. This process is further consolidated by the upregulation of ribosome biogenesis factors and the acquisition of the YAP/TAZ-dependent undifferentiated cell state. Finally, we show that RNA polymerase I is a therapeutic Achilles\' heel for both keratinocytes and OSCC patient-derived cells endowed with high VAV2 catalytic activity. Collectively, these findings highlight the therapeutic potential of modulating VAV2 and the ribosome biogenesis pathways in both preneoplastic and late progression stages of OSCC.
    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
    下一代雄激素受体信号抑制剂(ARSI),如恩扎鲁他胺(恩扎)和达鲁他胺(达罗),最初可有效治疗晚期前列腺癌(PCa)和去势抵抗前列腺癌(CRPC)。然而,患者经常复发并产生交叉抵抗,因此,耐药性成为CRPC相关死亡的必然原因。通过对GEO数据集进行全面分析,CRISPR全基因组筛选结果,ATAC-seq数据,和RNA-seq数据,由于PAK1/RELA/hnRNPA1/AR-V7轴的激活,我们系统确定PAK1是ARSI交叉耐药的重要因素.抑制PAK1随后抑制NF-κB途径和AR-V7表达可有效克服ARSI交叉抗性。我们的发现表明,PAK1代表了临床上治疗ARSI交叉耐药PCa患者的有希望的治疗靶基因。意义声明:PAK1在前列腺癌进展中驱动ARSI交叉耐药。
    Next-generation androgen receptor signaling inhibitors (ARSIs), such as enzalutamide (Enza) and darolutamide (Daro), are initially effective for the treatment of advanced prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). However, patients often relapse and develop cross-resistance, which consequently makes drug resistance an inevitable cause of CRPC-related mortality. By conducting a comprehensive analysis of GEO datasets, CRISPR genome-wide screening results, ATAC-seq data, and RNA-seq data, we systemically identified PAK1 as a significant contributor to ARSI cross-resistance due to the activation of the PAK1/RELA/hnRNPA1/AR-V7 axis. Inhibition of PAK1 followed by suppression of NF-κB pathways and AR-V7 expression effectively overcomes ARSI cross-resistance. Our findings indicate that PAK1 represents a promising therapeutic target gene for the treatment of ARSI cross-resistant PCa patients in the clinic. STATEMENT OF SIGNIFICANCE: PAK1 drives ARSI cross-resistance in prostate cancer progression.
    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