FAK

fak
  • 文章类型: Journal Article
    背景:先兆子痫与母体炎症过度反应和母胎界面免疫失衡有关。除免疫失衡外,最近还认识到促炎趋化因子Fractalkine(CX3CL1)。在这项研究中,初步探讨了CX3CL1-CX3C趋化因子受体1(CX3CR1)对子痫前期蜕膜巨噬细胞功能和滋养细胞侵袭能力的调控。
    方法:该研究包括60名妇女,这些妇女被分配到NP组(血压正常的孕妇,n=30)和sPE组(患有重度子痫前期的女性,n=30)。交货后,免疫组化法检测两组胎盘组织中CX3CL1的表达。通过WesternBlot检测胎盘组织中CX3CL1和蜕膜组织中CX3CR1的蛋白水平,并通过免疫荧光检测CX3CR1在蜕膜中的表达。巨噬细胞极化为经典活化的(M1)巨噬细胞。M1用PBS治疗(对照组),重组人CX3CL1(CX3CL1组),重组人CX3CL1+选择性CX3CR1拮抗剂-JMS-17-2(CX3CL1+抗CX3CR1组)和重组人CX3CL1+选择性CX3CR1拮抗剂-JMS-17-2+VS-6063(CX3CL1+抗CX3CR1+FAK抑制剂组)。如前所述共培养M1和HTR8/SVneo细胞以通过transwell测定和伤口愈合测定评估侵袭和迁移能力。
    结果:在这项研究中,CX3CL1在重度先兆子痫(sPE)患者胎盘组织中的表达高于正常血压妊娠(NP)。CX3CR1在重度子痫前期患者的蜕膜组织中高表达,主要在蜕膜组织的巨噬细胞中表达。CX3CL1/CX3CR1通过FAK信号通路降低M1巨噬细胞中VEGF的表达,并降低HTR-8/SVneo的侵袭和迁移功能。
    结论:这些发现揭示了CX3CL1-CX3CR1通过FAK调节滋养细胞功能,并为子痫前期的发病机制提供了新的见解。
    BACKGROUND: Preeclampsia is associated with maternal inflammatory overreaction and imbalanced immunity at the mother-fetus interface. The pro-inflammatory chemokine fractalkine (CX3CL1) is recently recognized apart from imbalanced immunity. In this study, CX3CL1- CX3C chemokine receptor 1(CX3CR1) regulation of decidual macrophage function and trophoblast invasion ability in preeclampsia was initially explored.
    METHODS: The study comprised 60 women allocated to NP group (normotensive pregnant woman, n = 30) and sPE group (woman with severe preeclampsia, n = 30). After the delivery, the expression of CX3CL1 in placental tissues of the two groups was detected by immunohistochemical analysis. The protein level of CX3CL1 in placental tissue and CX3CR1 in decidua tissue was detected by Western Blot and the localization of CX3CR1 expression in decidua was detected by immunofluorescence. Macrophages were polarized into classically activated (M1) macrophages. M1 were treat with PBS (control group), recombinant human CX3CL1 (CX3CL1 group), recombinant human CX3CL1+ selective CX3CR1 antagonist-JMS-17-2 (CX3CL1+anti-CX3CR1 group) and recombinant human CX3CL1 + selective CX3CR1 antagonist-JMS-17-2 + VS-6063 (CX3CL1+anti-CX3CR1+ FAK inhibitor group). M1 and HTR8/SVneo cells were co-cultured as described previously to assess invasion and migration capacity by transwell assays and Wound-healing assay.
    RESULTS: In this study, CX3CL1 expression is high in the placental tissues of severe preeclampsia (sPE) patients than in normotensive pregnancies (NP). CX3CR1 expression is high in the decidual tissues of severe preeclampsia patients and mainly expressed in macrophages of decidual tissues. CX3CL1/CX3CR1 decreased VEGF expression in M1 macrophages and reduced the invasion and migration function of HTR-8/SVneo through the FAK signaling pathway.
    CONCLUSIONS: These findings revealed that CX3CL1-CX3CR1 regulate the trophoblast function by FAK and provided new insights into the pathogenesis of preeclampsia.
    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:通过内体囊泡回收整合素对于癌细胞的迁移至关重要,这导致胰腺癌的转移和毁灭性的癌症相关的死亡。所以,需要开发新的诊断和治疗分子,靶向内体囊泡的再循环。
    方法:公共数据库,包括TCGA,ICGC,分析GSE21501、GSE28735和GENT以获得诊断和治疗靶标。为了揭示分子靶标的生物学作用和潜在机制,进行了各种分子生物学实验。
    结果:首先,我们确定了UNC13D在胰腺癌患者中的过度表达(n=824)及其在四个独立的胰腺癌队列中的预后意义和高风险比(HR)(TCGA,n=178,p=0.014,HR=3.629;ICGC,n=91,p=0.000,HR=4.362;GSE21501,n=102,p=0.002,HR=2.339;GSE28735,n=45,p=0.022,HR=2.681)。此外,其表达与胰腺癌的临床病理进展有关。进一步的生物学研究表明,UNC13D通过调节FAK磷酸化将再循环内体的胞吐作用与粘着灶性周转耦合来调节胰腺癌细胞的迁移。免疫沉淀和免疫细胞化学显示整合素再循环期间内体中RAB11-UNC13D-FAK轴的形成。我们观察到UNC13D直接与FAK的FERM结构域相互作用,并以钙依赖性方式调节FAK磷酸化。最后,我们发现UNC13D和FAK的共表达显示最差的存活率(TCGA,p=0.000;ICGC,p=0.036;GSE28735,p=0.006)。
    结论:我们强调UNC13D,一个新的预后因素,通过RAB11-UNC13D-FAK轴将整联蛋白再循环与局灶性粘附周转耦合以促进胰腺癌细胞的迁移,从而促进胰腺癌的进展。
    BACKGROUND: Recycling of integrin via endosomal vesicles is critical for the migration of cancer cells, which leads to the metastasis of pancreatic cancer and devastating cancer-related death. So, new diagnostic and therapeutic molecules which target the recycling of endosomal vesicles need to be developed.
    METHODS: Public databases including TCGA, ICGC, GSE21501, GSE28735, and GENT are analyzed to derive diagnostic and therapeutic targets. To reveal biological roles and underlying mechanisms of molecular targets, various molecular biological experiments were conducted.
    RESULTS: First, we identified UNC13D\'s overexpression in patients with pancreatic cancer (n = 824) and its prognostic significance and high hazard ratio (HR) in four independent pancreatic cancer cohorts (TCGA, n = 178, p = 0.014, HR = 3.629; ICGC, n = 91, p = 0.000, HR = 4.362; GSE21501, n = 102, p = 0.002, HR = 2.339; GSE28735, n = 45, p = 0.022, HR = 2.681). Additionally, its expression is associated with the clinicopathological progression of pancreatic cancer. Further biological studies have shown that UNC13D regulates the migration of pancreatic cancer cells by coupling the exocytosis of recycling endosomes with focal adhesion turnover via the regulation of FAK phosphorylation. Immunoprecipitation and immunocytochemistry showed the formation of the RAB11-UNC13D-FAK axis in endosomes during integrin recycling. We observed that UNC13D directly interacted with the FERM domain of FAK and regulated FAK phosphorylation in a calcium-dependent manner. Finally, we found co-expression of UNC13D and FAK showed the poorest survival (TCGA, p = 0.000; ICGC, p = 0.036; GSE28735, p = 0.006).
    CONCLUSIONS: We highlight that UNC13D, a novel prognostic factor, promotes pancreatic cancer progression by coupling integrin recycling with focal adhesion turnover via the RAB11-UNC13D-FAK axis for the migration of pancreatic cancer cells.
    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
    以前批准的药物的重新定位占据了研究人员的计划。巴氯芬(Bac)是我们确定的神经保护能力的候选人,提出了作为非离子表面活性剂基纳米囊泡(NISNV)配方的有效药物递送,以对抗大鼠轻度重复性创伤性脑损伤(mRTBI),从而减少口服或注射药物的数量,尤其是严重昏迷患者或儿科患者。
    A(23)阶乘设计用于限制Bac加载的NISNV公式,检查了一堆变量。进行体内实验以经皮测试制备的配方的功效。测量以下参数:γ-氨基丁酸B(GABAB)的脑表达,蛋白激酶C-α(PKC-α),粘着斑激酶(FAK),TNF-α和核因子κB(NF-κB)p65,丙二醛(MDA),超氧化物歧化酶(SOD),和组织病理学。
    前者的粒度(PS)和包封效率百分比(EE%)从60.40±0.28%散斑至88.02±0.01%,后者为174.64±0.93至1174.50±3.54nm。8小时后的体外释放%为63.25±5.47%至84.79±3.75%。优化的配方(F4)说明了可取性=1,在8小时内渗透了630.09±3.53µg/cm2的Bac,相当于100%的Bac。Bac创伤后治疗可恢复GABAB和PKC-α的脑表达,同时减少FAK。除了增强组织学发现,通过降低TNF-α和NF-κBp65,抗炎作用明显。因此,通过降低MDA水平和恢复SOD活性,本文揭示了显着的抗氧化后遗症。
    负载Bac的Niosome的透皮递送证实了神经保护作用,并成功地超越了皮肤到大脑的屏障,这使得它成为反复创伤的有希望的治疗选择。
    UNASSIGNED: The repositioning of previously approved drugs is occupying the researchers\' plans. Baclofen (Bac) was our candidate for its established neuroprotective capacity, with a proposal of efficient drug delivery as non-ionic surfactant-based nanovesicles (NISNV) formulae against mild repetitive traumatic brain injury (mRTBI) in rats, thus reducing the number of orally or injected medications, especially in severely comatose patients or pediatrics.
    UNASSIGNED: A (23) factorial design was implemented for confining Bac-loaded NISNV formulae, where a bunch of variables were inspected. An in-vivo experiment was done to test the prepared formula\'s efficacy transdermally. The following parameters were measured: brain expression of gamma amino butyric acid B (GABAB), protein kinase C- α (PKC-α), focal adhesion kinase (FAK), TNF-α and nuclear factor kappa B (NF-κB) p65, malondialdehyde (MDA), superoxide dismutase (SOD), and histopathology.
    UNASSIGNED: The particle size (PS) and entrapment efficiency percent (EE%) speckled from 60.40±0.28% to 88.02±0.01% for the former and 174.64±0.93 to 1174.50±3.54 nm for the latter. In vitro release% after 8 hours ranged from 63.25±5.47% to 84.79±3.75%. The optimized formula (F4) illustrated desirability=1, with 630.09±3.53 µg/cm2 of Bac permeated over 8 hours, which equates to 100% of Bac. Bac post-trauma treatment restored brain expression of GABAB and PKC-α, while decreasing FAK. Besides enhancing the histological findings, the anti-inflammatory effect was clear by decreasing TNF-α and NF-κB p65. Consequently, significant antioxidant sequelae were revealed herein by diminishing MDA levels and restoring SOD activity.
    UNASSIGNED: Transdermal delivery of Bac-loaded niosomes confirmed neuroprotection and succeeded in surpassing skin-to-brain barriers, which makes it a promising therapeutic option for repeated traumas.
    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
    蛋白酪氨酸激酶(PTKs)除了作为治疗许多人类疾病的治疗靶标外,还作为信号通路中的关键分子发挥作用。包括癌症.PTKs的特征在于它们磷酸化丝氨酸的能力,苏氨酸,或酪氨酸残基,从而可以快速且可逆地改变其蛋白质底物的功能,其形式为蛋白质确认和与蛋白质伴侣相互作用的亲和力的显着变化,以在正常和病理条件下驱动细胞功能。PTKs分为两组:一组代表酪氨酸激酶,而另一个包括丝氨酸/苏氨酸激酶的成员。酪氨酸激酶组被细分为亚组:其中一个包括受体酪氨酸激酶(RTK)的成员,而另一个亚组包括非受体酪氨酸激酶(NRTK)的成员。这两个激酶基团在许多细胞功能中都起着“开”或“关”开关的作用。NRTK是在许多癌症类型中过表达和活化的酶,并且响应于细胞外信号传导依赖性机制来调节可变的细胞功能。NRTK介导的不同细胞功能在细胞质或细胞核中受到激酶依赖性和激酶非依赖性机制的调节。因此,靶向NRTKs对于改善不同肿瘤类型的治疗策略具有重要意义。本文综述了NRTKs在肿瘤进展和耐药中的结构和机制作用及其作为肿瘤治疗靶点的重要性。
    Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an \"on\" or \"off\" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
    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
    肝细胞癌(HCC)是一个重要的全球健康问题,高发病率和死亡率。BucidarasinA,一种天然的二萜,已显示在一系列肿瘤细胞系中发挥显着的细胞毒性作用。然而,造成这种细胞毒性的潜在机制尚不清楚.在这项研究中,我们试图阐明bucidarasinA的抗肿瘤机制,一种天然的二萜,来自于Caseariagraveolens,特别关注其对肝癌的影响。此外,我们采用表面等离子体共振(SPR),分子对接,和细胞热转移测定(CETSA)以进一步了解bucidarasinA的靶蛋白。我们的发现表明bucidarasinA对HepG2细胞表现出明显的细胞毒性。体外分析表明,木耳苦艾素A在S期中断细胞周期,通过调节FAK和STAT3信号通路抑制HepG2细胞的增殖和转移。此外,体内研究表明,bucidarasinA不仅具有抗肿瘤作用,而且还阻碍了新生血管的形成,血管内皮生长因子(VEGF)和bucidarasinA之间的SPR相互作用证实了这一发现。这项研究证实了bucidarasinA,氯洛丹二萜,作为HCC的治疗候选药物,通过直接靶向STAT3和FAK信号传导途径,在体外和体内均显示出实质性的抗肿瘤功效。
    Hepatocellular carcinoma (HCC) is a significant global health concern, with high rates of morbidity and mortality. Bucidarasin A, a natural diterpenoid, has been shown to exert notable cytotoxic effects across a range of tumor cell lines. However, the underlying mechanisms responsible for this cytotoxicity remain unclear. In this study, we sought to elucidate the antitumor mechanisms of bucidarasin A, a natural diterpenoid derived from Casearia graveolens, with a particular focus on its effects on HCC. Furthermore, we employed surface plasmon resonance (SPR), molecular docking, and cellular thermal shift assay (CETSA) to gain further insight into the target protein of bucidarasin A. Our findings revealed that bucidarasin A exhibited pronounced cytotoxicity towards HepG2 cells. In vitro analysis indicated that bucidarasin A interrupted the cell cycle at the S phase and inhibited the proliferation and metastasis of HepG2 cells by modulating the FAK and STAT3 signaling pathways. Moreover, in vivo studies demonstrated that bucidarasin A not only exhibited antitumor effects but also impeded neovascularization, a finding that was corroborated by SPR interactions between vascular endothelial growth factor (VEGF) and bucidarasin A. This research substantiated that bucidarasin A, a clerodane diterpenoid, held promise as a therapeutic candidate against HCC, showcasing substantial antitumor efficacy both in vitro and in vivo through direct targeting of the STAT3 and FAK signaling pathways.
    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:增加的半乳糖凝集素3结合蛋白(LGALS3BP)血清水平已用于评估肝纤维化分期和肝细胞癌(HCC)的严重程度。考虑到转化生长因子-β1(TGF-β1)在这些疾病的出现中的关键作用,本研究验证了LGALS3BP调节TGF-β1信号通路的假设。
    方法:分析代谢功能障碍相关脂肪性肝炎(MASH)和HCC患者中LGALS3BP和TGFB1的表达水平。多种组学技术,比如RNA测序,转座酶可接近的染色质测序测定,和液相色谱-串联质谱蛋白质组学,用于鉴定LGALS3BP-TGF-β1轴的调节机制。在条件性LGALS3BP敲入和LGALS3BP敲除小鼠中研究了LGALS3BP改变的TGF-β1信号传导的作用。
    结果:在MASH和HCC患者中,LGALS3BP和TGFB1水平呈正相关。炎性细胞因子干扰素α刺激肝癌细胞中LGALS3BP或肝细胞中LGALS3BP的异位过表达促进了TGFB1的表达水平。在肝细胞特异性LGALS3BP敲入小鼠的肝脏中观察到加重的纤维化,TGFB1水平升高。LGALS3BP直接结合并组装整合素αV,从具有重排的F-肌动蛋白细胞骨架的细胞外潜伏复合物中释放活性TGF-β1所需的整合介质。释放的TGF-β1激活JunB转录因子,这反过来又促进了TGF-β1正反馈回路。肝细胞中LGALS3BP缺失下调TGF-β1信号和CCl4诱导的纤维化。此外,LGALS3BP耗竭通过限制纤维化TGF-β1的可用性来阻碍肝癌发生。
    结论:LGALS3BP通过控制TGF-β1信号通路在肝纤维化和癌变中起关键作用,使其成为TGF-β1相关疾病的有希望的治疗靶点。
    BACKGROUND: Increased Galectin 3-binding protein (LGALS3BP) serum levels have been used to assess hepatic fibrosis stages and the severity of hepatocellular carcinoma (HCC). Considering the crucial role of transforming growth factor-β1 (TGF-β1) in the emergence of these diseases, the present study tested the hypothesis that LGALS3BP regulates the TGF-β1 signaling pathway.
    METHODS: The expression levels of LGALS3BP and TGFB1 were analyzed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and HCC. Multiple omics techniques, such as RNA-sequencing, transposase-accessible chromatin-sequencing assay, and liquid chromatography-tandem mass spectrometry proteomics, were used to identify the regulatory mechanisms for the LGALS3BP-TGF-β1 axis. The effects of altered TGF-β1 signaling by LGALS3BP were investigated in conditional LGALS3BP-knockin and LGALS3BP-knockout mice.
    RESULTS: In patients with MASH and HCC, the levels of LGALS3BP and TGFB1 exhibited positive correlations. Stimulation of LGALS3BP by the inflammatory cytokine interferon α in HCC cells or ectopic overexpression of LGALS3BP in hepatocytes promoted the expression levels of TGFB1. Aggravated fibrosis was observed in the livers of hepatocyte-specific LGALS3BP-knockin mice, with increased TGFB1 levels. LGALS3BP directly bound to and assembled integrin αV, an integral mediator required for releasing active TGF-β1 from extracellular latent complex with the rearranged F-actin cytoskeleton. The released TGF-β1 activated JunB transcription factor, which in turn promoted the TGF-β1 positive feedback loop. LGALS3BP deletion in the hepatocytes downregulated TGF-β1 signaling and CCl4 induced fibrosis. Moreover, LGALS3BP depletion hindered hepatocarcinogenesis by limiting the availability of fibrogenic TGF-β1.
    CONCLUSIONS: LGALS3BP plays a crucial role in hepatic fibrosis and carcinogenesis by controlling the TGF-β1 signaling pathway, making it a promising therapeutic target in TGF-β1-related diseases.
    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
    机械刺激是细胞环境中的关键物理因素。机械传导是细胞行为的基本调节因子,调节细胞增殖,分化,凋亡,并在病理过程中表现出特定的特征改变。随着研究的继续,表观遗传科学在机械转导中的作用引起了人们的注意。然而,机械转导与表观遗传学在生理和病理过程中协同作用的分子机制尚未阐明。我们专注于组蛋白如何修饰,作为表观遗传学的重要组成部分,与多个信号通路协调以控制细胞命运和疾病进展。具体来说,我们认为组蛋白修饰可以与信号通路形成调节反馈回路,也就是说,组蛋白修饰不仅可以作为靶基因转录信号通路的下游调控因子,还可以为调控信号通路提供反馈。机械转导和表观遗传变化可能是临床实践中的潜在标志物和治疗靶标。
    Mechanical stimulation is the key physical factor in cell environment. Mechanotransduction acts as a fundamental regulator of cell behavior, regulating cell proliferation, differentiation, apoptosis, and exhibiting specific signature alterations during the pathological process. As research continues, the role of epigenetic science in mechanotransduction is attracting attention. However, the molecular mechanism of the synergistic effect between mechanotransduction and epigenetics in physiological and pathological processes has not been clarified. We focus on how histone modifications, as important components of epigenetics, are coordinated with multiple signaling pathways to control cell fate and disease progression. Specifically, we propose that histone modifications can form regulatory feedback loops with signaling pathways, that is, histone modifications can not only serve as downstream regulators of signaling pathways for target gene transcription but also provide feedback to regulate signaling pathways. Mechanotransduction and epigenetic changes could be potential markers and therapeutic targets in clinical practice.
    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
    背景:细胞外基质(ECM)硬度与糖尿病心肌病(DCM)的进展以及DCM治疗对抗糖尿病药物的反应密切相关。Dapagliflozin(Dapa)已被证明对糖尿病具有心脏保护作用,并被列为治疗心力衰竭的一线药物。但是ECM硬度与Dapa治疗效果之间的调节关系仍然难以捉摸。
    方法:这项工作使用体内DCM大鼠模型和具有高糖损伤的体外心肌细胞模型研究了ECM硬度对DCM进展和Dapa功效的影响。首先,通过不同程度的心肌损伤的DCM大鼠模型和Dapa治疗4周,心肌损伤的水平,心肌氧化应激,获得了AT1R(机械信号蛋白)的表达和心肌组织的硬度。然后,为了模仿DCM早期和晚期心肌组织的硬度,我们通过在具有两种硬度并暴露于高糖水平且没有/有Dapa干预的聚丙烯酰胺凝胶上培养H9c2心肌细胞来构建细胞模型。细胞活力,获得了活性氧(ROS)水平和机械信号敏感蛋白的表达。
    结果:DCM进展伴随着心肌组织僵硬度的增加,高糖可协同加重心肌细胞损伤。Dapa可以改善ECM刚度诱导的DCM进展,其对DCM的疗效在软ECM上更明显,与AT1R-FAK-NOX2的调控通路有关。此外,Dapa可抑制ECM诱导的整合素β1的表达,但对压电蛋白1无明显影响。
    结论:我们的研究发现生物力学在DCM进展中的调节和作用以及Dapa对DCM的疗效。为DCM治疗提供新的见解。此外,我们的工作表明早期Dapa干预下DCM的临床预后较好。
    BACKGROUND: Extracellular matrix (ECM) stiffness is closely related to the progress of diabetic cardiomyopathy (DCM) and the response of treatment of DCM to anti-diabetic drugs. Dapagliflozin (Dapa) has been proven to have cardio-protective efficacy for diabetes and listed as the first-line drug to treat heart failure. But the regulatory relationship between ECM stiffness and treatment efficacy of Dapa remains elusive.
    METHODS: This work investigated the effect of ECM stiffness on DCM progression and Dapa efficacy using both in vivo DCM rat model and in vitro myocardial cell model with high glucose injury. First, through DCM rat models with various levels of myocardial injury and administration with Dapa treatment for four weeks, the levels of myocardial injury, myocardial oxidative stress, expressions of AT1R (a mechanical signal protein) and the stiffness of myocardial tissues were obtained. Then for mimicking the stiffness of myocardial tissues at early and late stages of DCM, we constructed cell models through culturing H9c2 myocardial cells on the polyacrylamide gels with two stiffness and exposed to a high glucose level and without/with Dapa intervention. The cell viability, reactive oxygen species (ROS) levels and expressions of mechanical signal sensitive proteins were obtained.
    RESULTS: The DCM progression is accompanied by the increased myocardial tissue stiffness, which can synergistically exacerbate myocardial cell injury with high glucose. Dapa can improve the ECM stiffness-induced DCM progression and its efficacy on DCM is more pronounced on the soft ECM, which is related to the regulation pathway of AT1R-FAK-NOX2. Besides, Dapa can inhibit the expression of the ECM-induced integrin β1, but without significant impact on piezo 1.
    CONCLUSIONS: Our study found the regulation and effect of biomechanics in the DCM progression and on the Dapa efficacy on DCM, providing the new insights for the DCM treatment. Additionally, our work showed the better clinical prognosis of DCM under early Dapa intervention.
    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
    赖氨酰氧化酶(LOX)在高度僵硬的侵袭性肿瘤中上调,与转移相关,阻力,更糟糕的生存;然而,目前没有有效的,安全,和口服生物可利用的小分子LOX抑制剂在临床上治疗这些侵袭性增生性实体瘤。在这里,我们发现了双噻唑衍生物作为有效的LOX抑制剂,通过强有力的药物样分子筛选与基于细胞/重组蛋白的测定相结合。结构-活性关系分析确定了一种有效的先导化合物(LXG6403),与LOXL2相比,LOX具有3.5倍特异性,而不抑制LOXL1,时间和浓度依赖性的不可逆抑制模式。LXG6403显示出良好的药代动力学特性,全球改变ECM/胶原蛋白结构,并降低肿瘤硬度。这导致更好的药物渗透,抑制FAK信号,并诱导ROS/DNA损伤,G1逮捕,化疗耐药三阴性乳腺癌(TNBC)细胞系的凋亡,PDX类器官,和体内。总的来说,我们的有效和可耐受的双噻唑LOX抑制剂增强TNBC的化学反应,最致命的乳腺癌亚型.
    Lysyl oxidase (LOX) is upregulated in highly stiff aggressive tumors, correlating with metastasis, resistance, and worse survival; however, there are currently no potent, safe, and orally bioavailable small molecule LOX inhibitors to treat these aggressive desmoplastic solid tumors in clinics. Here we discovered bi-thiazole derivatives as potent LOX inhibitors by robust screening of drug-like molecules combined with cell/recombinant protein-based assays. Structure-activity relationship analysis identified a potent lead compound (LXG6403) with ∼3.5-fold specificity for LOX compared to LOXL2 while not inhibiting LOXL1 with a competitive, time- and concentration-dependent irreversible mode of inhibition. LXG6403 shows favorable pharmacokinetic properties, globally changes ECM/collagen architecture, and reduces tumor stiffness. This leads to better drug penetration, inhibits FAK signaling, and induces ROS/DNA damage, G1 arrest, and apoptosis in chemoresistant triple-negative breast cancer (TNBC) cell lines, PDX organoids, and in vivo. Overall, our potent and tolerable bi-thiazole LOX inhibitor enhances chemoresponse in TNBC, the deadliest breast cancer subtype.
    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
    骨骼肌构成了动物体内最大比例的组织,并且在生物体内正常生命活动的发展中起着关键作用。然而,骨骼肌生长发育的调控机制目前尚不清楚。本研究研究了Ankrd1对C2C12成肌细胞增殖和分化的影响。这里,我们确定Ankrd1是肌肉细胞发育的潜在调节因子,发现Ankrd1敲低导致C2C12细胞的增殖能力降低,但分化水平增加。基因本体论和京都百科全书的基因和基因组途径富集分析以及RNA-seq结果表明,Ankrd1敲低激活了粘着斑激酶(FAK)/F-肌动蛋白信号通路,大多数基因在该通路中显着富集上调。当Ankrd1敲低时,整合素亚基Itga6启动子活性增加,如通过双荧光素酶报告分析所证明的。这项研究揭示了Ankrd1敲低通过改变整合素亚基水平增强FAK磷酸化活性的分子机制。从而激活FAK/Rho-GTP酶/F-肌动蛋白信号通路,最终促进成肌细胞分化。我们的数据表明Ankrd1可能是肌肉细胞发育的潜在调节因子。我们的发现为骨骼肌的生长发育提供了新的见解,为进一步研究人类肌肉相关疾病提供了有价值的参考。
    Skeletal muscle constitutes the largest percentage of tissue in the animal body and plays a pivotal role in the development of normal life activities in the organism. However, the regulation mechanism of skeletal muscle growth and development remains largely unclear. This study investigated the effects of Ankrd1 on the proliferation and differentiation of C2C12 myoblasts. Here, we identified Ankrd1 as a potential regulator of muscle cell development, and found that Ankrd1 knockdown resulted in the proliferation ability decrease but the differentiation level increase of C2C12 cells. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyzes as well as RNA-seq results showed that Ankrd1 knockdown activated focal adhesion kinase (FAK)/F-actin signal pathway with most genes significantly enriched in this pathway upregulated. The integrin subunit Itga6 promoter activity is increased when Ankrd1 knockdown, as demonstrated by a dual-luciferase reporter assay. This study revealed the molecular mechanism by which Ankrd1 knockdown enhanced FAK phosphorylation activity through the alteration of integrin subunit levels, thus activating FAK/Rho-GTPase/F-actin signal pathway, eventually promoting myoblast differentiation. Our data suggested that Ankrd1 might serve as a potential regulator of muscle cell development. Our findings provide new insights into skeletal muscle growth and development and valuable references for further study of human muscle-related diseases.
    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