Proto-Oncogene Proteins c-pim-1

原癌基因蛋白质 c - pim - 1
  • 文章类型: Journal Article
    血小板产生的关键步骤是巨核细胞迁移到骨髓内的血管窦中。这种归巢是由趋化因子CXCL12及其受体CXCR4介导的。CXCR4也是血小板活化和血栓形成的正调节因子。Pim-1激酶已被证明在其他细胞类型中调节CXCR4信号传导,我们之前已经描述了Pim激酶抑制剂如何减弱血小板聚集至CXCL12。然而,Pim-1调节血小板和巨核细胞中CXCR4信号传导的机制尚未阐明。使用人类血小板,鼠骨髓来源的巨核细胞,和巨核细胞细胞系MEG-01,我们证明药理学Pim激酶抑制导致巨核细胞和血小板对CXCL12的功能反应减少,包括减少巨核细胞迁移和血小板颗粒分泌。发现CXCL12信号传导的减弱归因于CXCR4的表面表达降低。发现CXCR4表面水平的降低是由快速受体内化介导的,在没有激动剂刺激的情况下。我们证明,Pim激酶抑制药物通过减少组成型CXCR4表面表达破坏巨核细胞和血小板功能,减少可用于激动剂刺激和信号传导的受体的数量。这些发现对开发和使用Pim激酶抑制剂治疗与CXCL12/SDF1α循环水平升高和血栓形成风险增加相关的病症具有意义。
    A key step in platelet production is the migration of megakaryocytes to the vascular sinusoids within the bone marrow. This homing is mediated by the chemokine CXCL12 and its receptor CXCR4. CXCR4 is also a positive regulator of platelet activation and thrombosis. Pim-1 kinase has been shown to regulate CXCR4 signalling in other cell types, and we have previously described how Pim kinase inhibitors attenuate platelet aggregation to CXCL12. However, the mechanism by which Pim-1 regulates CXCR4 signalling in platelets and megakaryocytes has yet to be elucidated. Using human platelets, murine bone marrow-derived megakaryocytes, and the megakaryocyte cell line MEG-01, we demonstrate that pharmacological Pim kinase inhibition leads to reduced megakaryocyte and platelet function responses to CXCL12, including reduced megakaryocyte migration and platelet granule secretion. Attenuation of CXCL12 signalling was found to be attributed to the reduced surface expression of CXCR4. The decrease in CXCR4 surface levels was found to be mediated by rapid receptor internalisation, in the absence of agonist stimulation. We demonstrate that pharmacological Pim kinase inhibition disrupts megakaryocyte and platelet function by reducing constitutive CXCR4 surface expression, decreasing the number of receptors available for agonist stimulation and signalling. These findings have implications for the development and use of Pim kinase inhibitors for the treatment of conditions associated with elevated circulating levels of CXCL12/SDF1α and increased thrombotic risk.
    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
    肿瘤相关巨噬细胞(TAM)的高度浸润驱动肿瘤进展并与各种肿瘤类型的不良预后相关。我们的研究发现,在非小细胞肺癌(NSCLC)中Pim-1原癌基因(PIM1)的消融抑制了TAM浸润,并防止它们向M2表型极化。从而重塑肿瘤免疫微环境(TME)。PIM1对巨噬细胞趋化和极化的影响的主要机制涉及CC基序趋化因子配体2(CCL2)。非小细胞肺癌组织中PIM1的表达水平与CCL2的高表达呈正相关。导致患者总体生存率下降。机械上,PIM1缺陷通过靶向核因子κβ(NF-κB)信号促进TAM的重编程,并抑制NSCLC细胞的CCL2反式激活。CCL2分泌的减少阻碍了TAM的积累及其向肿瘤前表型的极化。此外,Pim1和PD-1的双重阻断协同抑制肿瘤生长,重新极化的巨噬细胞,并提高了抗PD-1抗体的疗效。总的来说,我们的研究结果阐明了PIM1在NSCLCTME内协调TAM中的关键作用,并强调了PIM1抑制作为增强癌症免疫治疗疗效的策略的潜力.
    Elevated infiltration of tumor-associated macrophages (TAMs) drives tumor progression and correlates with poor prognosis for various tumor types. Our research identifies that the ablation of the Pim-1 proto-oncogene (PIM1) in non-small cell lung cancer (NSCLC) suppresses TAM infiltration and prevents them from polarizing toward the M2 phenotype, thereby reshaping the tumor immune microenvironment (TME). The predominant mechanism through which PIM1 exerts its impact on macrophage chemotaxis and polarization involves CC motif chemokine ligand 2 (CCL2). The expression level of PIM1 is positively correlated with high CCL2 expression in NSCLC, conferring a worse overall patient survival. Mechanistically, PIM1 deficiency facilitates the reprogramming of TAMs by targeting nuclear factor kappa beta (NF-κB) signaling and inhibits CCL2 transactivation by NSCLC cells. The decreased secretion of CCL2 impedes TAM accumulation and their polarization toward a pro-tumoral phenotype. Furthermore, Dual blockade of Pim1 and PD-1 collaboratively suppressed tumor growth, repolarized macrophages, and boosted the efficacy of anti-PD-1 antibody. Collectively, our findings elucidate the pivotal role of PIM1 in orchestrating TAMs within the TME of NSCLC and highlight the potential of PIM1 inhibition as a strategy for enhancing the efficacy of cancer immunotherapy.
    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
    丝氨酸/苏氨酸蛋白激酶(CK2,PIM-1,RIO1)具有组成活性,高度保守,多效性,和多功能激酶,控制几个信号通路并调节许多细胞功能,如细胞活动,生存,扩散,和凋亡。在过去的几十年里,它们作为潜在的治疗靶点越来越受到关注,从各种癌症和神经系统,炎症,自身免疫性疾病和病毒性疾病,包括COVID-19。尽管积累了大量的实验数据,仍然没有“配方”可以促进寻找新的有效激酶抑制剂。我们研究的目的是开发一种有效的筛选方法,可用于此目的。结合密度泛函理论计算和分子对接,补充了新开发的定量方法,用于比较结合模式,提供了对一组导致其抑制作用的理想特性的深入了解。数学度量有助于评估结合模式之间的距离,而热图揭示了配体中应该根据结合位点要求进行修饰的位置。本文提出的结构结合亲和力指数和结构结合亲和力景观有助于测量响应于配体结构中相对较小的变化而获得或失去结合亲和力的程度。物理化学概况与上述因素的结合使得能够识别“死亡”和“有希望”搜索方向。对实验数据进行的测试已经验证并证明了所提出的创新方法的高效率。我们量化配体之间的差异及其结合能力的方法有望指导未来对新抗癌剂的研究。
    Serine/threonine protein kinases (CK2, PIM-1, RIO1) are constitutively active, highly conserved, pleiotropic, and multifunctional kinases, which control several signaling pathways and regulate many cellular functions, such as cell activity, survival, proliferation, and apoptosis. Over the past decades, they have gained increasing attention as potential therapeutic targets, ranging from various cancers and neurological, inflammation, and autoimmune disorders to viral diseases, including COVID-19. Despite the accumulation of a vast amount of experimental data, there is still no \"recipe\" that would facilitate the search for new effective kinase inhibitors. The aim of our study was to develop an effective screening method that would be useful for this purpose. A combination of Density Functional Theory calculations and molecular docking, supplemented with newly developed quantitative methods for the comparison of the binding modes, provided deep insight into the set of desirable properties responsible for their inhibition. The mathematical metrics helped assess the distance between the binding modes, while heatmaps revealed the locations in the ligand that should be modified according to binding site requirements. The Structure-Binding Affinity Index and Structural-Binding Affinity Landscape proposed in this paper helped to measure the extent to which binding affinity is gained or lost in response to a relatively small change in the ligand\'s structure. The combination of the physico-chemical profile with the aforementioned factors enabled the identification of both \"dead\" and \"promising\" search directions. Tests carried out on experimental data have validated and demonstrated the high efficiency of the proposed innovative approach. Our method for quantifying differences between the ligands and their binding capabilities holds promise for guiding future research on new anti-cancer agents.
    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:核孔蛋白98(NUP98)融合蛋白在白血病中反复发现,并与不利的临床结果相关。它们分布到细胞核,并通过异常的转录调节促进白血病的发生。我们先前使用下一代测序在T细胞急性淋巴细胞白血病(T-ALL)患者中鉴定了NUP98-BPTF(NB)融合。NUP98的FG-重复和PHD指和溴结构域PHD指转录因子(BPTF)的溴结构域保留在融合中。像其他NUP98融合蛋白一样,NB被认为是调节白血病发生所必需的基因。然而,其靶基因或途径仍然未知。
    方法:为了研究NB融合蛋白的潜在致癌特性,我们将强力霉素诱导的NB表达载体慢病毒转导至小鼠NIH3T3成纤维细胞和人JurkatT-ALL细胞.
    结果:NB通过上调编码丝氨酸/苏氨酸激酶的原癌基因Pim1促进小鼠NIH3T3成纤维细胞的转化。NB通过与其启动子结合并激活MYC和mTORC1信号传导来转录调节Pim1表达。PIM1敲低或mTORC1信号传导的药理学抑制抑制NB诱导的NIH3T3细胞转化。此外,NB通过失活促凋亡蛋白BCL2相关的细胞死亡激动剂(BAD)来增强人JurkatT-ALL细胞的存活。
    结论:我们证明了NB在细胞转化和存活中的关键作用,并将PIM1确定为NB的关键下游靶标。这些发现为NB融合阳性白血病患者提出了有希望的治疗策略。
    BACKGROUND: Nucleoporin 98 (NUP98) fusion proteins are recurrently found in leukemia and are associated with unfavorable clinical outcomes. They are distributed to the nucleus and contribute to leukemogenesis via aberrant transcriptional regulation. We previously identified NUP98-BPTF (NB) fusion in patients with T-cell acute lymphoblastic leukemia (T-ALL) using next-generation sequencing. The FG-repeat of NUP98 and the PHD finger and bromodomain of bromodomain PHD finger transcription factor (BPTF) are retained in the fusion. Like other NUP98 fusion proteins, NB is considered to regulate genes that are essential for leukemogenesis. However, its target genes or pathways remain unknown.
    METHODS: To investigate the potential oncogenic properties of the NB fusion protein, we lentivirally transduced a doxycycline-inducible NB expression vector into mouse NIH3T3 fibroblasts and human Jurkat T-ALL cells.
    RESULTS: NB promoted the transformation of mouse NIH3T3 fibroblasts by upregulating the proto-oncogene Pim1, which encodes a serine/threonine kinase. NB transcriptionally regulated Pim1 expression by binding to its promoter and activated MYC and mTORC1 signaling. PIM1 knockdown or pharmacological inhibition of mTORC1 signaling suppressed NB-induced NIH3T3 cell transformation. Furthermore, NB enhanced the survival of human Jurkat T-ALL cells by inactivating the pro-apoptotic protein BCL2-associated agonist of cell death (BAD).
    CONCLUSIONS: We demonstrated the pivotal role of NB in cell transformation and survival and identified PIM1as a key downstream target of NB. These findings propose a promising therapeutic strategy for patients with NB fusion-positive leukemia.
    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
    由于其简单性,CRISPR-Cas9系统已成为最流行的基因编辑技术,效率高,和低成本。然而,在该系统中,同源定向修复(HDR)介导的基因敲入效率低,这限制了其在动物模型制备中的应用,基因治疗,农业遗传改良。这里,我们报告了一种简单有效的基于报告子的检测方法的设计和优化,以可视化和量化HDR效率.通过随机筛选一个小分子化合物库,两组化合物,包括拓扑异构酶抑制剂和PIM1激酶抑制剂,已被确定为促进HDR。两个代表性化合物,依托泊苷和槲皮素,也显著提高CRISPR-Cas9和HDR介导的小鼠胚胎基因敲入的效率。我们的研究不仅提供了筛选可能促进HDR的化合物的测定法,而且还鉴定了有用的工具化合物,以促进使用CRISPR-Cas9系统构建转基因动物模型。
    The CRISPR-Cas9 system has emerged as the most prevalent gene editing technology due to its simplicity, high efficiency, and low cost. However, the homology-directed repair (HDR)-mediated gene knock-in in this system suffers from low efficiency, which limits its application in animal model preparation, gene therapy, and agricultural genetic improvement. Here, we report the design and optimization of a simple and efficient reporter-based assay to visualize and quantify HDR efficiency. Through random screening of a small molecule compound library, two groups of compounds, including the topoisomerase inhibitors and PIM1 kinase inhibitors, have been identified to promote HDR. Two representative compounds, etoposide and quercetagetin, also significantly enhance the efficiency of CRISPR-Cas9 and HDR-mediated gene knock-in in mouse embryos. Our study not only provides an assay to screen compounds that may facilitate HDR but also identifies useful tool compounds to facilitate the construction of genetically modified animal models with the CRISPR-Cas9 system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    OBJECTIVE: To investigate the effects of the serine/threonine kinase family member 1 (PIM1) gene on the proliferation and apoptosis of acute myeloid leukemia (AML) U937 cells, and the regulation effect on Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway.
    METHODS: Bone marrow mononuclear cells from newly diagnosed adult AML patients and patients with iron deficiency anemia were collected and PIM1 mRNA expression was detected by RT-qPCR. AML cell line U937 cells were divided into U937 group (U937 cells were cultured normally), Si-PIM1 group (U937 cells were transfected with low expression adenovirus vector containing PIM1 mRNA), Si-NC group (U937 cells were transfected with low expression adenovirus vector without PIM1 mRNA), coumermycin A1 (CoA1) group (JAK2 activator CoA1 was added to U937 cells at a concentration of 20 μmol/L), and Si-PIM1+CoA1 group (U937 cells were transfected with adenoviral vector containing low expression of PIM1 mRNA and added with CoA1 at a concentration of 20 μmol/L). After culture for 24 h, the expressions of PIM1 mRNA and protein, JAK2/STAT3 pathway, cell cycle and apoptosis-related proteins in U937 cells were detected by RT-qPCR and Western blot, the cell proliferation activity was detected by MTT assay, and flow cytometry was used to detect cell cycle changes and apoptosis rate.
    RESULTS: The PIM1 mRNA expression level in bone marrow mononuclear cells in AML patients was higher than that in patients with iron deficiency anemia (P < 0.05). Compared with U937 group, PIM1 mRNA and protein, phosphorylated JAK2 (p-JAK2)/JAK2, phosphorylated STAT3 (p-STAT3)/STAT3, Cyclin D1, cyclin-dependent kinase 2 (CDK2) protein, cell proliferation activity, S phase and G 2/M phase proportions were decreased in Si-PIM1 group (all P < 0.05), while p27, Caspase-3 protein, G0/G1 phase proportion and apoptosis rate were increased (all P < 0.05). However, the changes of above indicators in CoA1 group were just opposite to those in Si-PIM1 group, indicating that CoA1 could reverse the effect of Si-PIM1 on U937 cells. There were no significant differences in above indexes of U937 cells between U937 group, Si-PIM1+CoA1 group and Si-NC group (P >0.05).
    CONCLUSIONS: Knockdown of PIM1 gene expression can inhibit U937 cell proliferation and promote apoptosis, in order to alleviate ALM process, which may be related to the inhibition of JAK2/STAT3 pathway activation.
    UNASSIGNED: PIM1基因对急性髓系白血病U937细胞增殖、凋亡及JAK2/STAT3信号通路的影响.
    UNASSIGNED: 探讨PIM1基因对急性髓系白血病(AML)U937细胞增殖、凋亡的影响,以及对JAK2/STAT3通路的调控作用。.
    UNASSIGNED: 收集初诊成人AML患者和单纯缺铁性贫血患者的骨髓单个核细胞,荧光定量PCR检测PIM1 mRNA表达。将AML细胞系U937细胞分为:U937组(U937细胞正常培养)、Si-PIM1组(U937细胞转染含PIM1 mRNA的低表达腺病毒载体)、Si-NC组(U937细胞转染不含PIM1 mRNA的低表达腺病毒载体)、CoA1组(U937细胞中加入浓度为20 μmol/L的JAK2激活剂CoA1)、Si-PIM1+CoA1组(U937细胞转染含PIM1 mRNA低表达的腺病毒载体并加入浓度为20 μmol/L的CoA1)。培养24 h。荧光定量PCR和蛋白印迹法检测U937细胞PIM1 mRNA和蛋白、JAK2/STAT3通路、细胞周期、凋亡相关蛋白表达;噻唑蓝法检测细胞增殖活性;流式细胞术检测细胞周期变化及凋亡率。.
    UNASSIGNED: AML患者骨髓单个核细胞中PIM1 mRNA表达水平高于单纯缺铁性贫血患者(P < 0.05)。与U937组相比,Si-PIM1组细胞PIM1 mRNA和蛋白、p-JAK2/JAK2、p-STAT3/STAT3、Cyclin D1、CDK2蛋白、细胞增殖活性、S期比例、G2/M期比例降低(均P < 0.05),p27、Caspase-3蛋白、G0/G1期、凋亡率升高(均P < 0.05),而CoA1组上述指标的变化情况与Si-PIM1组正好相反,CoA1可逆转Si-PIM1对U937细胞的作用效果。U937组、Si-PIM1+CoA1组、Si-NC组U937细胞上述指标差异无统计学意义(P >0.05)。.
    UNASSIGNED: 敲低PIM1基因表达可抑制U937细胞增殖、促进凋亡,缓解ALM进程,且上述作用可能与抑制JAK2/STAT3通路活化有关。.
    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
    据报道,PIM激酶(PIM-1,PIM-2和PIM-3)在控制细胞存活的信号级联中起关键作用,扩散,和差异化。这些激酶的过度表达导致血液恶性肿瘤,如弥漫性大B细胞淋巴瘤(DLBCL),多发性骨髓瘤,白血病,淋巴瘤和前列腺癌等.PIM激酶作为生物标志物和潜在的治疗靶标已显示出对精确癌症治疗的希望。选择性PIM-1、PIM-2和/或PIM-3同种型抑制剂已经在患有晚期癌症(包括复发性/难治性癌症)的患者中显示出显著结果。
    关于PIM激酶(PIM-1,PIM-2和PIM-3)在肿瘤发生中的综合文献综述,获得专利的PIM激酶抑制剂(2016年至今),和他们的药理学和结构见解已被强调。
    最近,PIM激酶即.作为治疗靶标的PIM-1、PIM-2和PIM-3(丝氨酸/苏氨酸蛋白激酶家族的成员)已经在肿瘤学特别是血液恶性肿瘤中引起了相当大的兴趣。专利的PIM激酶抑制剂由杂环(稠合)环结构(如吲哚)组成,吡啶,吡嗪,吡唑,吖嗪,哌嗪,噻唑,恶二唑,喹啉,三唑并吡啶,吡唑并吡啶,咪唑并吡啶嗪,恶二唑-硫酮,吡唑并嘧啶,三唑并吡啶嗪,咪唑并吡啶嗪,吡唑并喹唑啉和吡唑并吡啶等。在癌症化疗中显示出有希望的结果。
    UNASSIGNED: PIM Kinases (PIM-1, PIM-2, and PIM-3) have been reported to play crucial role in signaling cascades that govern cell survival, proliferation, and differentiation. Over-expression of these kinases leads to hematological malignancies such as diffuse large B cell lymphomas (DLBCL), multiple myeloma, leukemia, lymphoma and prostate cancer etc. PIM kinases as biomarkers and potential therapeutic targets have shown promise toward precision cancer therapy. The selective PIM-1, PIM-2, and/or PIM-3 isoform inhibitors have shown significant results in patients with advanced stages of cancer including relapsed/refractory cancer.
    UNASSIGNED: A comprehensive literature review of PIM Kinases (PIM-1, PIM-2, and PIM-3) in oncogenesis, the patented PIM kinase inhibitors (2016-Present), and their pharmacological and structural insights have been highlighted.
    UNASSIGNED: Recently, PIM kinases viz. PIM-1, PIM-2, and PIM-3 (members of the serine/threonine protein kinase family) as therapeutic targets have attracted considerable interest in oncology especially in hematological malignancies. The patented PIM kinase inhibitors comprised of heterocyclic (fused)ring structure(s) like indole, pyridine, pyrazine, pyrazole, pyridazine, piperazine, thiazole, oxadiazole, quinoline, triazolo-pyridine, pyrazolo-pyridine, imidazo-pyridazine, oxadiazole-thione, pyrazolo-pyrimidine, triazolo-pyridazine, imidazo-pyridazine, pyrazolo-quinazoline and pyrazolo-pyridine etc. showed promising results in cancer chemotherapy.
    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
    Moloney鼠白血病病毒(PIM)家族丝氨酸/苏氨酸激酶的前病毒整合位点通过磷酸化参与肿瘤代谢的底物,在血液恶性肿瘤和实体瘤中发挥促致瘤功能,细胞存活,转移,炎症,和免疫细胞入侵。然而,目前缺乏对PIM激酶功能的全面了解.目前正在评估多种小分子PIM激酶抑制剂作为癌症患者的共治疗剂。为了进一步阐明PIM激酶在癌症中的功能,我们使用反向凝胶内激酶测定法对PIM1底物进行了深度分析,以鉴定可被小分子靶向的下游细胞过程.假定的PIM底物提名的RNA剪接和rRNA加工作为PIM调节的细胞过程的途径分析。PIM抑制引起PIM抑制剂反应性急性髓性白血病(AML)细胞系的可重复剪接变化。PIM抑制剂与针对剪接因子3b亚基1(SF3B1)和丝氨酸-精氨酸蛋白激酶1(SRPK1)的剪接调节剂协同作用以杀死AML细胞。PIM抑制也改变了rRNA加工,和PIM抑制剂与RNA聚合酶I抑制剂协同杀死AML细胞并阻断AML肿瘤生长。这些数据表明,深入的激酶底物知识可以阐明未被理解的激酶功能,提名协同联合治疗策略。这种方法可能会扩展联合治疗设备,以克服激酶抑制剂抗性疾病,该疾病会限制恶性疾病的持久反应。
    UNASSIGNED: Provirus integration site for Moloney murine leukemia virus (PIM) family serine/threonine kinases perform protumorigenic functions in hematologic malignancies and solid tumors by phosphorylating substrates involved in tumor metabolism, cell survival, metastasis, inflammation, and immune cell invasion. However, a comprehensive understanding of PIM kinase functions is currently lacking. Multiple small-molecule PIM kinase inhibitors are currently being evaluated as cotherapeutics in patients with cancer. To further illuminate PIM kinase functions in cancer, we deeply profiled PIM1 substrates using the reverse in-gel kinase assay to identify downstream cellular processes targetable with small molecules. Pathway analyses of putative PIM substrates nominated RNA splicing and ribosomal RNA (rRNA) processing as PIM-regulated cellular processes. PIM inhibition elicited reproducible splicing changes in PIM-inhibitor-responsive acute myeloid leukemia (AML) cell lines. PIM inhibitors synergized with splicing modulators targeting splicing factor 3b subunit 1 (SF3B1) and serine-arginine protein kinase 1 (SRPK1) to kill AML cells. PIM inhibition also altered rRNA processing, and PIM inhibitors synergized with an RNA polymerase I inhibitor to kill AML cells and block AML tumor growth. These data demonstrate that deep kinase substrate knowledge can illuminate unappreciated kinase functions, nominating synergistic cotherapeutic strategies. This approach may expand the cotherapeutic armamentarium to overcome kinase inhibitor-resistant disease that limits durable responses in malignant disease.
    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
    目的:非酒精性脂肪性肝病(NAFLD)已成为全球重大公共卫生问题,其中垂直袖状胃切除术(VSG)已成为一种有效的治疗方法。该研究试图阐明PIM1缓解NAFLD进展的过程。莫洛尼鼠白血病病毒1(PIM1)的亲病毒整合位点起丝氨酸/苏氨酸激酶的作用。生物信息学分析显示,NAFLD中PIM1的表达降低。
    方法:为了进一步证明PIM1在NAFLD中的作用,进行了深入的体内实验,其中雄性C57BL/6小鼠随机分组接受正常或高脂饮食19周。对它们进行操作或递送PIM1过表达的负载的腺相关病毒(AAV-PIM1)。在体外实验中,用棕榈酸处理AML12细胞以诱导肝脂肪变性。
    结果:结果显示VSG手术和病毒递送小鼠减轻了氧化应激,和体内凋亡。对于AML12细胞,氧化应激水平,凋亡,脂质代谢通过PIM1上调而降低。此外,ML385处理导致NRF2/HO-1/NQO1信号级联的下调,表明PIM1通过靶向该途径减轻NAFLD。
    结论:PIM1通过调节NRF2/HO-1/NQO1信号通路减轻高脂饮食诱导的小鼠肝脏氧化应激和NAFLD。
    OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD) has risen as a significant global public health issue, for which vertical sleeve gastrectomy (VSG) has become an effective treatment method. The study sought to elucidate the processes through which PIM1 mitigates the advancement of NAFLD. The Pro-viral integration site for Moloney murine leukemia virus 1 (PIM1) functions as a serine/threonine kinase. Bioinformatics analysis revealed that reduced PIM1 expression in NAFLD.
    METHODS: To further prove the role of PIM1 in NAFLD, an in-depth in vivo experiment was performed, in which male C57BL/6 mice were randomly grouped to receive a normal or high-fat diet for 24 weeks. They were operated or delivered the loaded adeno-associated virus which the PIM1 was overexpressed (AAV-PIM1). In an in vitro experiment, AML12 cells were treated with palmitic acid to induce hepatic steatosis.
    RESULTS: The results revealed that the VSG surgery and virus delivery of mice alleviated oxidative stress, and apoptosis in vivo. For AML12 cells, the levels of oxidative stress, apoptosis, and lipid metabolism were reduced via PIM1 upregulation. Moreover, ML385 treatment resulted in the downregulation of the NRF2/HO-1/NQO1 signaling cascade, indicating that PIM1 mitigates NAFLD by targeting this pathway.
    CONCLUSIONS: PIM1 alleviated mice liver oxidative stress and NAFLD induced by high-fat diet by regulating the NRF2/HO-1/NQO1 signaling Pathway.
    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
    莫洛尼鼠白血病病毒(PIM)激酶的前整合,属于丝氨酸/苏氨酸激酶家族,已经发现在各种类型的癌症中过度表达,比如前列腺,乳房,结肠,子宫内膜,胃,还有胰腺癌.三种同工型PIM激酶,即,PIM1,PIM2和PIM3共享高度的序列和结构相似性,并磷酸化控制肿瘤表型如增殖和细胞存活的底物。靶向短寿命的PIM激酶提出了一个有趣的策略,因为体内敲除研究导致非致死表型。表明PIM的临床抑制可能具有较少的不良反应。ATP结合位点(铰链区)具有独特的属性,这导致了针对一种或所有三种PIM同工型的新型小分子支架的开发。机器学习和基于结构的方法一直处于在临床前和临床环境中开发针对PIM的新型有效化学疗法的最前沿。还没有人获得癌症治疗的批准。PIM同工型的稳定性由PIM激酶活性维持,这导致对PIM抑制剂和化疗的耐药性;因此,为了克服这种影响,目前正在开发特异性降解PIM蛋白的PIM蛋白水解靶向嵌合体(PROTACs)。在这次审查中,我们概述了PIM激酶的致癌功能,他们的结构,函数,以及不同类型癌症的关键信号网络,和药理学小分子抑制剂的潜力。Further,我们的全面综述还为未来开发特异性靶向PIM激酶的新型抗肿瘤药物提供了有价值的见解.总之,我们提供了有关降解PIM激酶的好处的见解,而不是阻断其催化活性,以解决PIM激酶的致癌潜力。
    The proviral integration for the Moloney murine leukemia virus (PIM) kinases, belonging to serine/threonine kinase family, have been found to be overexpressed in various types of cancers, such as prostate, breast, colon, endometrial, gastric, and pancreatic cancer. The three isoforms PIM kinases i.e., PIM1, PIM2, and PIM3 share a high degree of sequence and structural similarity and phosphorylate substrates controlling tumorigenic phenotypes like proliferation and cell survival. Targeting short-lived PIM kinases presents an intriguing strategy as in vivo knock-down studies result in non-lethal phenotypes, indicating that clinical inhibition of PIM might have fewer adverse effects. The ATP binding site (hinge region) possesses distinctive attributes, which led to the development of novel small molecule scaffolds that target either one or all three PIM isoforms. Machine learning and structure-based approaches have been at the forefront of developing novel and effective chemical therapeutics against PIM in preclinical and clinical settings, and none have yet received approval for cancer treatment. The stability of PIM isoforms is maintained by PIM kinase activity, which leads to resistance against PIM inhibitors and chemotherapy; thus, to overcome such effects, PIM proteolysis targeting chimeras (PROTACs) are now being developed that specifically degrade PIM proteins. In this review, we recapitulate an overview of the oncogenic functions of PIM kinases, their structure, function, and crucial signaling network in different types of cancer, and the potential of pharmacological small-molecule inhibitors. Further, our comprehensive review also provides valuable insights for developing novel antitumor drugs that specifically target PIM kinases in the future. In conclusion, we provide insights into the benefits of degrading PIM kinases as opposed to blocking their catalytic activity to address the oncogenic potential of PIM kinases.
    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