MEK1/2

MEK1 / 2
  • 文章类型: Journal Article
    听力损失是最常见的残疾类型之一;然而,只有一种FDA批准的药物可以预防任何类型的听力损失。用高效化疗药物治疗,顺铂,和暴露于高分贝噪声是听力损失的两个最常见的原因。丝裂原活化蛋白激酶(MAPK)途径,由RAF组成的磷酸化级联,MEK1/2和ERK1/2与两种类型的听力损失有关。药理学抑制BRAF或ERK1/2在多种小鼠模型中对噪声和顺铂诱导的听力损失具有保护作用。曲美替尼,MEK1/2抑制剂,保护小鼠耳蜗外植体免受顺铂诱导的外毛细胞死亡;然而,据我们所知,抑制MEK1/2尚未显示在体内对听力损失有保护作用。在这项研究中,我们证明,在翻译相关小鼠模型中,曲美替尼可以预防顺铂诱导的听力损失,并且不会干扰顺铂在癌细胞系中的肿瘤杀伤功效.高剂量的曲美替尼与顺铂合用时对小鼠有毒性,但较低剂量的药物对听力损失有保护作用,没有任何已知的毒性。曲美替尼还保护小鼠免受噪声引起的听力损失和突触损伤。这项研究表明,MEK1/2抑制可以防止听力损失的两种损害,以及靶向MAPK途径中的所有三种激酶可保护小鼠免受顺铂和噪声引起的听力损失。
    Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high-decibel noises are two of the most common causes of hearing loss. The mitogen-activated protein kinase (MAPK) pathway, a phosphorylation cascade consisting of RAF, MEK1/2, and ERK1/2, has been implicated in both types of hearing loss. Pharmacologically inhibiting BRAF or ERK1/2 is protective against noise- and cisplatin-induced hearing loss in multiple mouse models. Trametinib, a MEK1/2 inhibitor, protects from cisplatin-induced outer hair cell death in mouse cochlear explants; however, to the best of our knowledge, inhibiting MEK1/2 has not yet been shown to be protective against hearing loss in vivo. In this study, we demonstrate that trametinib protects against cisplatin-induced hearing loss in a translationally relevant mouse model and does not interfere with cisplatin\'s tumor-killing efficacy in cancer cell lines. Higher doses of trametinib were toxic to mice when combined with cisplatin, but lower doses of the drug were protective against hearing loss without any known toxicity. Trametinib also protected mice from noise-induced hearing loss and synaptic damage. This study shows that MEK1/2 inhibition protects against both insults of hearing loss, as well as that targeting all three kinases in the MAPK pathway protects mice from cisplatin- and noise-induced hearing loss.
    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
    听力损失是最常见的残疾类型之一;然而,只有一种FDA批准的药物可以预防任何类型的听力损失。用高效化疗药物治疗,顺铂,和暴露于高分贝噪声是听力损失的两个最常见的原因。丝裂原活化蛋白激酶(MAPK)途径,由RAF组成的磷酸化级联,MEK1/2和ERK1/2与两种类型的听力损失有关。药理学抑制BRAF或ERK1/2在多种小鼠模型中对噪声和顺铂诱导的听力损失具有保护作用。曲美替尼,MEK1/2抑制剂,保护小鼠耳蜗外植体免受顺铂诱导的外毛细胞死亡;然而,据我们所知,抑制MEK1/2尚未显示在体内对听力损失有保护作用。在这项研究中,我们证明,在翻译相关小鼠模型中,曲美替尼可以防止顺铂诱导的听力损失,并且不会干扰顺铂在癌细胞系中的肿瘤杀伤功效.当与顺铂联合使用时,较高剂量的曲美替尼对小鼠有毒,但较低剂量的药物对听力损失具有保护作用,没有任何已知的毒性。曲美替尼还保护小鼠免受噪声引起的听力损失和突触损伤。这项研究表明,MEK1/2抑制可以保护小鼠免受听力损失的损害,并且靶向MAPK途径中的所有三种激酶可以保护小鼠免受顺铂和噪声引起的听力损失。
    Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high decibel noises are two of the most common causes of hearing loss. The mitogen activated protein kinase (MAPK) pathway, a phosphorylation cascade consisting of RAF, MEK1/2, and ERK1/2, has been implicated in both types of hearing loss. Pharmacologically inhibiting BRAF or ERK1/2 is protective from noise and cisplatin-induced hearing loss in multiple mouse models. Trametinib, a MEK1/2 inhibitor, protects from cisplatin induced outer hair cell death in mouse cochlear explants; however, to the best of our knowledge, inhibiting MEK1/2 has not yet been shown to be protective from hearing loss in vivo. In this study, we demonstrate that trametinib protects from cisplatin-induced hearing loss in a translationally relevant mouse model and does not interfere with cisplatin\'s tumor killing efficacy in cancer cell lines. Higher doses of trametinib were toxic to mice when combined with cisplatin but lower doses of the drug were protective from hearing loss without any known toxicity. Trametinib also protected mice from noise-induced hearing loss and synaptic damage. This study shows that MEK1/2 inhibition protects from both insults of hearing loss and that targeting all three kinases in the MAPK pathway protect from cisplatin and noise-induced hearing loss in mice.
    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
    临床前研究很少测试男女治疗的疗效。肿瘤学领域在这方面也不例外。在同基因模型中,原位,转移的胰腺导管腺癌,我们评估了性别对这种疾病的病理特征的影响,以及对一种新型的疗效和可能的不良副作用,在雄性和雌性C57BL/6J小鼠中,基于小分子的疗法抑制KRAS:SOS1,MEK1/2和PI3K信号传导。雄性小鼠的CD8阳性细胞肿瘤浸润较少,发展更大的肿瘤,与雌性小鼠相比,肺转移更多,存活概率更低。在实验结束时,雄性动物的这些更严重的病理特征伴随着更高的痛苦。评估的抑制剂BI-3406、曲美替尼和BKM120在体外显示出协同作用。这种组合疗法更有效地降低了雄性动物的肿瘤重量,尽管男女肿瘤中的药物浓度相似。这些结果强调了性别特异性临床前研究的重要性,同时为将来使用测试化合物的研究提供了坚实的基础。
    Preclinical studies rarely test the efficacy of therapies in both sexes. The field of oncology is no exception in this regard. In a model of syngeneic, orthotopic, metastasized pancreatic ductal adenocarcinoma we evaluated the impact of sex on pathological features of this disease as well as on the efficacy and possible adverse side effects of a novel, small molecule-based therapy inhibiting KRAS:SOS1, MEK1/2 and PI3K signaling in male and female C57BL/6J mice. Male mice had less tumor infiltration of CD8-positive cells, developed bigger tumors, had more lung metastasis and a lower probability of survival compared to female mice. These more severe pathological features in male animals were accompanied by higher distress at the end of the experiment. The evaluated inhibitors BI-3406, trametinib and BKM120 showed synergistic effects in vitro. This combinatorial therapy reduced tumor weight more efficiently in male animals, although the drug concentrations were similar in the tumors of both sexes. These results underline the importance of sex-specific preclinical research and at the same time provide a solid basis for future studies with the tested compounds.
    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:食管鳞状细胞癌(ESCC)是世界上消化系统肿瘤相关死亡的主要原因之一。不幸的是,在临床实践中,ESCC患者缺乏有效的化学预防剂,导致极高的死亡率。
    方法:筛选处方药物库,以发现ESCC细胞中的关键抗肿瘤特性。磷酸蛋白质组学,激酶阵列,应用下拉测定和药物亲和反应靶标稳定测定(DARTS)来探索机制并搜索协同靶标。建立的PDX小鼠模型用于确定多潘立酮的治疗效果。
    结果:在筛选处方药库之后,我们发现多潘立酮具有抗肿瘤特性。多潘立酮,作为胃动力剂,已广泛用于临床胃肠动力障碍。尽管研究有限,有迹象表明多潘立酮可能具有抗肿瘤特性。在这项研究中,我们确定多潘立酮在体外和体内均显着抑制ESCC的增殖。我们使用磷酸化蛋白质组学来揭示p-ERK,多潘立酮治疗后p-SMAD3下调。然后,激酶测定和下拉测定的结果进一步验证了多潘立酮与MEK1/2和CDK4直接结合,导致其激酶活性受到抑制。此外,我们的结果表明,MEK/ERK和CDK4/SMAD3信号通路是多潘立酮抗ESCC的主要通路。
    结论:总的来说,这些研究结果表明,多潘立酮是MEK1/2和CDK4的有效"多靶点"抑制剂,为ESCC的化学预防提供了潜在的益处.
    BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of digestive system tumor related death in the world. Unfortunately, effective chemopreventive agent is lack for patients with ESCC in clinical practice, which leads to the extremely high mortality rate.
    METHODS: A library of prescribed drugs was screened for finding critical anti-tumor properties in ESCC cells. The phosphoproteomics, kinase array, pulldown assay and drug affinity responsive target stabilization assay (DARTS) were applied to explore mechanisms and searched for synergistic targets. Established models of PDX in mice were used to determine the therapeutic effect of domperidone.
    RESULTS: After screening a library of prescribed drugs, we discovered that domperidone has anti-tumor properties. Domperidone, acting as a gastroprokinetic agent, has been widely used in clinic for gastrointestinal motility disorders. Despite limited research, there are indications that domperidone may have anti-tumor properties. In this study, we determined that domperidone significantly inhibited ESCC proliferation in vitro and in vivo. We employed phosphoproteomics to reveal p-ERK, and p-SMAD3 down-regulation upon domperidone treatment. Then, the results of kinase assay and pulldown assay further validated that domperidone directly combined with MEK1/2 and CDK4, leading to the inhibition of their kinase activity. Furthermore, our results revealed that MEK/ERK and CDK4/SMAD3 signal pathway were major pathways in domperidone against ESCC.
    CONCLUSIONS: Collectively, these findings suggest that domperidone serves as an effective \"multi-target\" inhibitor of MEK1/2 and CDK4, offering potential benefits for the chemoprevention of ESCC.
    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
    RAS调节的RAF-MEK1/2-ERK1/2信号通路在癌症中由于RAS蛋白(尤其是KRAS)的突变而被激活。BRAF,CRAF,MEK1和MEK2。虽然KRASG12C(肺腺癌)和BRAF和MEK1/2(黑色素瘤和结直肠癌)的抑制剂已获得临床批准,获得的抵抗力仍然是一个问题。因此,寻找新的抑制剂(尤其是RAS蛋白),该途径的新抑制剂模式和调节剂,这可能是新的药物靶点,继续并越来越多地涉及基于细胞的小分子筛选或遗传筛选,如RNAi,CRISPR或蛋白质干扰。在这里,我们描述了表现出多西环素依赖性表达KRASG12V或BRAFV600E的细胞系,并具有稳定整合的EGR1:EmGFP报告基因,可通过流式细胞术检测,高含量显微镜或免疫印迹。KRASG12V或BRAFV600E驱动的EmGFP表达被MEK1/2或ERK1/2抑制剂(MEKi和ERKi)抑制。BRAFi抑制BRAFV600E驱动的EmGFP表达,但增强对KRASG12V的反应,概括野生型RAF蛋白的矛盾激活。除了小分子,编码RAS特异性抗体片段的iDab6的表达抑制了KRASG12V-但不抑制BRAFV600E驱动的EmGFP表达。最后,EmGFP取代细菌硝基还原酶基因使KRASG12V或BRAFV600E在前药存在下驱动细胞死亡,这可能允许选择促进存活的途径抑制剂。这些细胞系应被证明可用于基于细胞的筛选,以鉴定KRAS或BRAF依赖性ERK1/2信号(药物靶标发现)的新调节因子,以及从药物发现筛选中筛选或分类“命中”。
    The RAS-regulated RAF-MEK1/2-ERK1/2 signalling pathway is activated in cancer due to mutations in RAS proteins (especially KRAS), BRAF, CRAF, MEK1 and MEK2. Whilst inhibitors of KRASG12C (lung adenocarcinoma) and BRAF and MEK1/2 (melanoma and colorectal cancer) are clinically approved, acquired resistance remains a problem. Consequently, the search for new inhibitors (especially of RAS proteins), new inhibitor modalities and regulators of this pathway, which may be new drug targets, continues and increasingly involves cell-based screens with small molecules or genetic screens such as RNAi, CRISPR or protein interference. Here we describe cell lines that exhibit doxycycline-dependent expression KRASG12V or BRAFV600E and harbour a stably integrated EGR1:EmGFP reporter gene that can be detected by flow cytometry, high-content microscopy or immunoblotting. KRASG12V or BRAFV600E-driven EmGFP expression is inhibited by MEK1/2 or ERK1/2 inhibitors (MEKi and ERKi). BRAFi inhibit BRAFV600E-driven EmGFP expression but enhance the response to KRASG12V, recapitulating paradoxical activation of wild type RAF proteins. In addition to small molecules, expression of iDab6, encoding a RAS-specific antibody fragment inhibited KRASG12V- but not BRAFV600E-driven EmGFP expression. Finally, substitution of EmGFP for a bacterial nitroreductase gene allowed KRASG12V or BRAFV600E to drive cell death in the presence of a pro-drug, which may allow selection of pathway inhibitors that promote survival. These cell lines should prove useful for cell-based screens to identify new regulators of KRAS- or BRAF-dependent ERK1/2 signalling (drug target discovery) as well as screening or triaging \'hits\' from drug discovery screens.
    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:种系分化中断或睾丸发育受损可导致不育或不育,并与人类睾丸癌症密切相关。在老鼠身上,SRY和SOX9诱导Fgf9表达,促进支持细胞分化和睾丸发育。FGF9也被认为促进雄性种系分化,但其机制未知。FGF通常通过丝裂原激活的蛋白激酶(MAPK)发出信号以磷酸化ERK1/2(pERK1/2)。我们探讨了FGF9是否通过在性腺性别确定和睾丸形成后立即抑制胎儿睾丸中的FGF或MEK1/2信号通过MAPK调节男性种系发育。但是在男性生殖系承诺之前。
    结果:在睾丸支持细胞中检测到pERK1/2,MEK1/2的抑制降低了睾丸支持细胞的增殖和组织,并导致一些生殖细胞位于睾丸外。虽然在生殖细胞中未检测到pERK1/2,体细胞性别决定严重破坏生殖细胞有丝分裂阻滞后MEK1/2的抑制,调节了广泛的男性种系发育基因,并阻止了关键男性种系标记的上调,DPPA4和DNMT3L。相比之下,而FGF抑制减少支持细胞增殖,雄性种系标记物的表达不受影响,生殖细胞正常进入有丝分裂停滞。虽然雄性种系分化没有被FGF抑制破坏,一系列干细胞和癌症相关基因通常在FGF或MEK1/2抑制24小时后发生改变,包括维持生殖干细胞的基因,节点信号,扩散,和种系癌症。
    结论:一起,这些数据证明了MEK1/2信号在睾丸发育过程中的新作用,这对于雄性种系分化至关重要,但表明对FGF信号传导的作用更有限。我们的数据表明,其他配体可能通过MEK1/2起作用以促进雄性种系分化,并强调需要对雄性种系发育进行进一步的机械理解。
    Disrupted germline differentiation or compromised testis development can lead to subfertility or infertility and are strongly associated with testis cancer in humans. In mice, SRY and SOX9 induce expression of Fgf9, which promotes Sertoli cell differentiation and testis development. FGF9 is also thought to promote male germline differentiation but the mechanism is unknown. FGFs typically signal through mitogen-activated protein kinases (MAPKs) to phosphorylate ERK1/2 (pERK1/2). We explored whether FGF9 regulates male germline development through MAPK by inhibiting either FGF or MEK1/2 signalling in the foetal testis immediately after gonadal sex determination and testis cord formation, but prior to male germline commitment.
    pERK1/2 was detected in Sertoli cells and inhibition of MEK1/2 reduced Sertoli cell proliferation and organisation and resulted in some germ cells localised outside of the testis cords. While pERK1/2 was not detected in germ cells, inhibition of MEK1/2 after somatic sex determination profoundly disrupted germ cell mitotic arrest, dysregulated a broad range of male germline development genes and prevented the upregulation of key male germline markers, DPPA4 and DNMT3L. In contrast, while FGF inhibition reduced Sertoli cell proliferation, expression of male germline markers was unaffected and germ cells entered mitotic arrest normally. While male germline differentiation was not disrupted by FGF inhibition, a range of stem cell and cancer-associated genes were commonly altered after 24 h of FGF or MEK1/2 inhibition, including genes involved in the maintenance of germline stem cells, Nodal signalling, proliferation, and germline cancer.
    Together, these data demonstrate a novel role for MEK1/2 signalling during testis development that is essential for male germline differentiation, but indicate a more limited role for FGF signalling. Our data indicate that additional ligands are likely to act through MEK1/2 to promote male germline differentiation and highlight a need for further mechanistic understanding of male germline development.
    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
    巨噬细胞在激活时经历不同的细胞状态,其可以对随后的刺激是低反应的(耐受的)或高反应的(引发的或训练的)。已知表观遗传修饰在确定这些细胞状态中起关键作用。然而,对导致这些表观遗传修饰的信号通路的作用知之甚少。这里,我们研究了针对脂多糖(LPS)诱导的关键信号通路的各种抑制剂对小鼠巨噬细胞耐受和启动的影响.我们发现,丝裂原活化蛋白激酶(MEK)1/2-细胞外信号调节激酶(ERK)1/2信号轴的长时间抑制(>18h)逆转了表达白细胞介素(IL)-1β和其他炎性细胞因子,如IL-6,肿瘤坏死因子(TNF)α,和CXCL10。催化活性和非活性MEK1突变体的异位表达抑制和增强了IL-1β的表达,分别。转录组学分析表明,由MEK1/2抑制剂U0126引发的细胞表达了更高水平的与免疫反应和细胞因子/趋化因子产生相关的基因集,但是随着细胞周期的进展,基因的表达水平较低,染色体组织,和异染色质形成比未引发的细胞。感兴趣的,组蛋白3赖氨酸9(H3K9)甲基转移酶Suv39h1和H3K9甲基化读数器Cbx5的mRNA表达被显著抑制,而H3K9脱甲基酶Kdm7a增强,提示MEK1/2-ERK信号轴在H3K9去甲基化中的作用。IL-1β基因组区域的H3K9三甲基化水平,TNFα,CXCL10降低了U0126。此外,H3K9甲基转移酶抑制剂BIX01294模拟了U0126的训练效应,而染色体盒同源物(CBX)5的过表达阻止了U0126在RAW264.7细胞和骨髓源性巨噬细胞中的训练效应.总的来说,这些数据表明,MEK1/2-ERK信号轴的长期抑制逆转了耐受,并可能通过降低H3K9甲基化水平引发巨噬细胞.
    Macrophages undergo different cellular states upon activation that can be hyporesponsive (tolerated) or hyperresponsive (primed or trained) to subsequent stimuli. Epigenetic modifications are known to play key roles in determining these cellular states. However, little is known about the role of signaling pathways that lead to these epigenetic modifications. Here, we examined the effects of various inhibitors targeting key signaling pathways induced by lipopolysaccharide (LPS) on tolerance and priming in murine macrophages. We found that a prolonged inhibition (>18 h) of the mitogen-activated protein kinase (MEK)1/2-extracellular signal-regulated kinase (ERK)1/2 signaling axis reversed tolerance and primed cells in expressing interleukin (IL)-1β and other inflammatory cytokines such as IL-6, tumor necrosis factor (TNF)α, and CXCL10. The ectopic expression of catalytically active and inactive MEK1 mutants suppressed and enhanced IL-1β expression, respectively. A transcriptomic analysis showed that cells primed by the MEK1/2 inhibitor U0126 expressed higher levels of gene sets associated with immune responses and cytokine/chemokine production, but expressed lower levels of genes with cell cycle progression, chromosome organization, and heterochromatin formation than non-primed cells. Of interest, the mRNA expressions of the histone 3 lysine 9 (H3K9) methyltransferase Suv39h1 and the H3K9 methylation reader Cbx5 were substantially suppressed, whereas the H3K9 demethylase Kdm7a was enhanced, suggesting a role of the MEK1/2-ERK signaling axis in H3K9 demethylation. The H3K9 trimethylation levels in the genomic regions of IL-1β, TNFα, and CXCL10 were decreased by U0126. Also, the H3K9 methyltransferase inhibitor BIX01294 mimicked the U0126 training effects and the overexpression of chromobox homolog (CBX)5 prevented the U0126 training effects in both RAW264.7 cells and bone-marrow-derived macrophages. Collectively, these data suggest that the prolonged inhibition of the MEK1/2-ERK signaling axis reverses tolerance and primed macrophages likely through decreasing the H3K9 methylation levels.
    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
    胶质母细胞瘤(GBM)仍然是一种无法治愈的疾病,五年复发率极高。我们研究了响应HDAC抑制(HDACi)与MEK1/2抑制(MEKi)或BCL-2家族抑制剂的神经胶质瘤干细胞(GSCs)凋亡。MEKi有效结合HDACi抑制生长,诱导细胞周期缺陷,和细胞凋亡,以及挽救促凋亡BH3蛋白BIM和BMF的表达。GSC的RNAseq分析揭示HDACi抑制促存活BCL-2家族基因MCL1和BCL-XL。因此,我们用BCL-2家族抑制剂代替MEKi,并观察到细胞凋亡增强。相反,癌症干细胞受体CD44的配体导致BMF减少,BIM,和凋亡。我们的数据强烈支持HDACi与MEKi或BCL-2家族抑制剂联合在神经胶质瘤中的进一步测试。
    Glioblastoma (GBM) remains an incurable disease with an extremely high five-year recurrence rate. We studied apoptosis in glioma stem cells (GSCs) in response to HDAC inhibition (HDACi) combined with MEK1/2 inhibition (MEKi) or BCL-2 family inhibitors. MEKi effectively combined with HDACi to suppress growth, induce cell cycle defects, and apoptosis, as well as to rescue the expression of the pro-apoptotic BH3-only proteins BIM and BMF. A RNAseq analysis of GSCs revealed that HDACi repressed the pro-survival BCL-2 family genes MCL1 and BCL-XL. We therefore replaced MEKi with BCL-2 family inhibitors and observed enhanced apoptosis. Conversely, a ligand for the cancer stem cell receptor CD44 led to reductions in BMF, BIM, and apoptosis. Our data strongly support further testing of HDACi in combination with MEKi or BCL-2 family inhibitors in glioma.
    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
    压力会引发可卡因使用的复发,这些可卡因参与了与记忆相关的细胞核的活动,如基底外侧杏仁核(BLA)和齿状回(DG)。临床前研究表明,D3受体(D3R)拮抗剂可能是减轻可卡因奖励和复发的有希望的手段。由于D3R调节Akt/mTOR和MEK/ERK1/2通路的活性,我们评估了SB-277011-A的效果,一个D3R拮抗剂,在恢复可卡因诱导的条件位置偏爱(CPP)过程中,这些激酶的活性由心理(约束)和生理(尾巴夹)应激引起。两种刺激都重新激活了熄灭的可卡因-CPP,但是只有受约束的动物在恢复期间会降低其运动活动。可卡因寻求行为再激活与p-Akt降低相关,p-mTOR,和p-ERK1/2在受约束动物的两个细胞核中的激活。虽然D3R阻断阻止了应激诱导的CPP恢复和血浆皮质酮增强,SB-277011-A明显调制的Akt,mTOR,和ERK1/2激活取决于应激源和使用的剂量。我们的数据支持皮质酮参与受限动物的SB-277011-A效应。此外,应激诱导复发期间BLA中p-mTOR/mTOR和/或p-ERK1/2/ERK1/2的比值似乎与接受48mg/kg拮抗剂的动物的运动活动有关.因此,我们的研究表明,D3R拮抗剂通过对记忆处理核中Akt/mTOR和MEK/ERK1/2通路的不同调节,可有效预防药物使用中应激性复发.
    Stress triggers relapses in cocaine use that engage the activity of memory-related nuclei, such as the basolateral amygdala (BLA) and dentate gyrus (DG). Preclinical research suggests that D3 receptor (D3R) antagonists may be a promising means to attenuate cocaine reward and relapse. As D3R regulates the activity of the Akt/mTOR and MEK/ERK1/2 pathways, we assessed the effects of SB-277011-A, a D3R antagonist, on the activity of these kinases during the reinstatement of cocaine-induced conditioned place preference (CPP) induced by psychological (restraint) and physiological (tail pinch) stress. Both stimuli reactivated an extinguished cocaine-CPP, but only restrained animals decreased their locomotor activity during reinstatement. Cocaine-seeking behavior reactivation was correlated with decreased p-Akt, p-mTOR, and p-ERK1/2 activation in both nuclei of restrained animals. While a D3R blockade prevented stress-induced CPP reinstatement and plasma corticosterone enhancement, SB-277011-A distinctly modulated Akt, mTOR, and ERK1/2 activation depending on the stressor and the dose used. Our data support the involvement of corticosterone in the SB-277011-A effects in restrained animals. Additionally, the ratios p-mTOR/mTOR and/or p-ERK1/2 /ERK1/2 in the BLA during stress-induced relapse seem to be related to the locomotor activity of animals receiving 48 mg/kg of the antagonist. Hence, our study indicates the D3R antagonist\'s efficacy to prevent stress-induced relapses in drug use through distinct modulation of Akt/mTOR and MEK/ERK1/2 pathways in memory-processing nuclei.
    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
    智人2号染色体克隆RP11-339H12(AC010883.5)是一种失调的长非编码RNA(lncRNA),从未在宫颈癌(CC)中进行过研究。因此,潜在的功能和分子机制尚不清楚。我们的研究探索了AC010883.5的生物学功能,以确定CC的潜在机制,并为改善临床治疗策略提供潜在的治疗靶点。我们使用定量实时聚合酶链反应来测量线粒体RNA水平,并使用蛋白质印迹来测量靶基因的蛋白质水平。Further,我们使用细胞计数试剂盒-8和5-溴-2'-脱氧尿苷掺入测定法来评估体外细胞增殖。通过流式细胞术分析细胞凋亡。通过伤口愈合分析细胞侵袭,并使用Transwell迁移测定法分析细胞迁移。最后,通过体内异种移植实验评估AC010883.5在CC生长中的生物学功能和机制。AC010883.5在CC组织和细胞系中增强,和增强AC010883.5的表达加速CC细胞增殖,迁移,以及在体外和体内的侵袭和诱导的上皮-间质转化。AC010883.5还通过促进细胞外信号调节激酶1/2的磷酸化来激活丝裂原活化蛋白激酶(MAPK)信号通路(即ERK1/2)和MAPK激酶1/2(即MEK1/2)。阻断MAPK信号通路可以抵消促增殖,亲移民,AC010883.5过表达的促侵袭作用。我们发现lncRNA,AC010883.5是通过MAPK信号通路失调参与CC肿瘤进展的致癌分子,这意味着AC010883.5可能是肿瘤进展和治疗反应的生物标志物。
    Homo sapiens chromosome 2 clone RP11-339H12 (AC010883.5) is a dysregulated long non-coding RNA (lncRNA) that has never been investigated in cervical cancer (CC). Thus, the potential function and molecular mechanism remain unclear. Our study explored the biological function of AC010883.5 to determine the underlying mechanisms in CC and provide potential therapeutic targets for improving the clinical treatment strategy. We used quantitative real-time polymerase chain reaction to measure mitochondrial RNA levels and western blot to measure the protein levels of target genes. Further, we used Cell Counting Kit-8 and 5-Bromo-2\'-deoxyuridine incorporation assays to evaluate cell proliferation in vitro. Cell apoptosis was analyzed by flow cytometry. Cell invasion was analyzed by wound healing and Transwell migration assays was ued to analyze cell migration. Finally, the biological function and mechanism of AC010883.5 in CC growth were evaluated by in vivo xenograft assay. AC010883.5 was enhanced in CC tissues and cell lines, and enhanced AC010883.5 expression accelerated CC cell proliferation, migration, and invasion and induced epithelial-mesenchymal transition in vitro and in vivo. AC010883.5 also activated the mitogen-activated protein kinase (MAPK) signaling pathway by promoting phosphorylation of extracellular signal-regulated kinase 1/2 (i.e., ERK1/2) and MAPK kinase 1/2 (i.e., MEK1/2). Blocking the MAPK signaling pathway could counteract the pro-proliferative, pro-migrative, and pro-invasive effects of AC010883.5 over-expression. We found that the lncRNA, AC010883.5, is an oncogenic molecule involved in CC tumor progression via dysregulation of the MAPK signaling pathway, implying that AC010883.5 could be a tumor progression and therapeutic response biomarker.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号