LIFR

LIFR
  • 文章类型: Journal Article
    Stüve-Wiedemann综合征(SWS),一种罕见的常染色体隐性疾病,以尺寸小巧为特征,细长骨头的曲率,弯曲的手指,体温升高的事件,呼吸窘迫或屏气,以及喂养方面的挑战,尤其是导致婴儿死亡。SWS是白血病抑制因子受体基因中潜在错义突变的结果,反映为蛋白质水平的许多氨基酸突变。在计算机工具和技术中使用Pred_MutHTP进行突变筛选,I-Mutant2.0,PANTHER.db,PolyPhen,将突变归类为有害/不稳定,结合实验数据分析,P136A和S279P作为“效应”引起的突变出现。预先存在的知识表明,SWS进展是构象改变和功能失调的LIFR,无法与LIF结合并进一步形成LIF/LIFR/gp130信号传导复合物。为了获得对所述突变对野生型蛋白质的影响的功能见解,一个全原子,明确,溶剂分子动力学模拟是按照对接方法进行的。因此,提到RMSD,RMSF,蛋白质动态网络分析,能量景观地块和域运动分析,揭示了未结合的LIFR_WT像往常一样更倾向于LIF结合,而突变体表现出相当大的结构域封闭以抑制LIF结合。我们通过MM/GBSA进行了结合亲和力分析,并在LIFR-LIF对接后进行了解离常数估计,发现与与SWS相关的柔性突变复合物相比,WT_复合物整体上更稳定和紧凑。我们的研究为了解LIFR突变的分子水平含义提供了途径,这为治疗干预开辟了途径。
    Stüve-Wiedemann syndrome (SWS), a rare autosomal recessive disorder, characterized by diminutive size, curvature of the elongated bones, bent fingers, episodes of heightened body temperature, respiratory distress or periods of breath-holding, and challenges with feeding, especially causes fatality in infants. SWS is an outcome of potential missense mutations in the leukemia inhibitory factor receptor gene reflected as numerous amino acid mutations at protein level. Employing in silico tools and techniques like mutational screening with Pred_MutHTP, I-Mutant2.0, PANTHER.db, PolyPhen, to classify mutations as deleterious/destabilizing, in conjunction with experimental data analysis, P136A and S279P emerged as \'effect\'-causing mutations. Pre-existing knowledge suggests, SWS progression is effectuated conformationally altered and dysfunctional LIFR, unable to bind to LIF and further form the LIF/LIFR/gp130 signalling complex. To gain functional insights into the effect of the said mutations on the wild type protein, an all-atom, explicit, solvent molecular dynamics simulation was performed following docking approaches. Consequently, referring to the RMSD, RMSF, protein dynamic network analysis, energy landscape plots and domain motion analysis, it was revealed that unbound LIFR_WT was more prone to LIF binding as usual whereas the mutants exhibited considerable domain closure to inhibit LIF binding. We conducted binding affinity analysis via MM/GBSA and dissociation constant estimation after LIFR-LIF docking and found the WT_complex to be more stable and compact as a whole when compared to the flexible mutant complexes thus being associated with SWS. Our study offers a route for understanding molecular level implications upon LIFR mutations which opens an avenue for therapeutic interventions.
    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
    放射治疗对于治疗结直肠癌(CRC)至关重要,尤其是晚期直肠癌.然而,CRC细胞的低放射敏感性极大地限制了放疗疗效。小核仁RNA(snoRNAs)是一类非编码RNA,主要指导核糖体RNA(rRNAs)的转录后修饰,小核RNA(snRNA),和其他细胞RNA。虽然snoRNAs参与肿瘤进展和化疗耐药,它们与放射敏感性的关联在很大程度上仍然未知.在这里,SNORA28在CRC中高表达,与不良预后呈正相关。此外,SNORA28过表达在体外和体内增强CRC细胞的生长和放射抗性。机械上,SNORA28充当分子诱饵,募集含溴结构域的蛋白4(BRD4),这增加了LIFR启动子区的H3K9乙酰化水平。这刺激LIFR转录,进而触发JAK1/STAT3通路,增强CRC细胞的增殖和放射抗性。总的来说,这些结果突出了snoRNAs调节肿瘤细胞放射敏感性和影响靶基因启动子区组蛋白乙酰化修饰的能力,从而拓宽了目前对snoRNA生物学功能和靶基因调控机制的认识。
    Radiotherapy is essential for treating colorectal cancer (CRC), especially in advanced rectal cancer. However, the low radiosensitivity of CRC cells greatly limits radiotherapy efficacy. Small nucleolar RNAs (snoRNAs) are a class of noncoding RNA that primarily direct post-transcriptional modifications of ribosomal RNAs (rRNAs), small nuclear RNAs (snRNAs), and other cellular RNAs. While snoRNAs are involved in tumor progression and chemoresistance, their association with radiosensitivity remains largely unknown. Herein, SNORA28 is shown highly expressed in CRC and is positively associated with poor prognosis. Furthermore, SNORA28 overexpression enhances the growth and radioresistance of CRC cells in vitro and in vivo. Mechanistically, SNORA28 acts as a molecular decoy that recruits bromodomain-containing protein 4 (BRD4), which increases the level of H3K9 acetylation at the LIFR promoter region. This stimulates LIFR transcription, which in turn triggers the JAK1/STAT3 pathway, enhancing the proliferation and radioresistance of CRC cells. Overall, these results highlight the ability of snoRNAs to regulate radiosensitivity in tumor cells and affect histone acetylation modification in the promoter region of target genes, thus broadening the current knowledge of snoRNA biological functions and the mechanism underlying target gene regulation.
    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
    睫状神经营养因子(CNTF)通过非信号α受体CNTF受体(CNTFR)和两种信号β受体糖蛋白130(gp130)和白血病抑制因子受体(LIFR)激活细胞。CNTF衍生的,Axokine对肥胖和胰岛素抵抗具有保护作用,但由于CNTF抗体的出现而停止了临床开发。嵌合细胞因子IC7使用具有来自CNTF的LIFR结合位点的白细胞介素(IL-)6的框架通过IL-6R:gp130:LIFR复合物激活细胞。类似于CNTF/Axokine,IC7保护小鼠免受肥胖和胰岛素抵抗。这里,我们开发了特异性靶向IL-6R:gp130:LIFR复合物的不依赖CNTF的嵌合体。在GIL-6和GIO-6中,我们分别将LIFR结合位点从LIF或OSM转移到IL-6。虽然GIO-6通过gp130:IL-6R:LIFR和gp130:IL-6R:OSMR复合物发出信号,GIL-6选择性激活IL-6R:gp130:LIFR受体复合物。通过对IC7和CNTF的重新评估,我们发现制瘤素M受体(OSMR)作为替代的非规范高亲和力受体,导致IL-6R:OSMR:gp130和CNTFR:OSMR:gp130受体复合物,分别。OSMR作为IC7和CNTF的替代高亲和力受体的发现表明GIL-6是第一个真正选择性的IL-6R:gp130:LIFR细胞因子,而GIO-6是IC7的无CNTF替代品。
    Ciliary neurotrophic factor (CNTF) activates cells via the non-signaling α-receptor CNTF receptor (CNTFR) and the two signaling β-receptors glycoprotein 130 (gp130) and leukemia inhibitory factor receptor (LIFR). The CNTF derivate, Axokine, was protective against obesity and insulin resistance, but clinical development was halted by the emergence of CNTF antibodies. The chimeric cytokine IC7 used the framework of interleukin (IL-)6 with the LIFR-binding site from CNTF to activate cells via IL-6R:gp130:LIFR complexes. Similar to CNTF/Axokine, IC7 protected mice from obesity and insulin resistance. Here, we developed CNTF-independent chimeras that specifically target the IL-6R:gp130:LIFR complex. In GIL-6 and GIO-6, we transferred the LIFR binding site from LIF or OSM to IL-6, respectively. While GIO-6 signals via gp130:IL-6R:LIFR and gp130:IL-6R:OSMR complexes, GIL-6 selectively activates the IL-6R:gp130:LIFR receptor complex. By re-evaluation of IC7 and CNTF, we discovered the Oncostatin M receptor (OSMR) as an alternative non-canonical high-affinity receptor leading to IL-6R:OSMR:gp130 and CNTFR:OSMR:gp130 receptor complexes, respectively. The discovery of OSMR as an alternative high-affinity receptor for IC7 and CNTF designates GIL-6 as the first truly selective IL-6R:gp130:LIFR cytokine, whereas GIO-6 is a CNTF-free alternative for IC7.
    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
    在以前的队列中已经观察到循环炎症蛋白与COVID-19之间的关系。然而,目前尚不清楚哪些循环炎症蛋白可能会增加COVID-19的风险或对COVID-19产生保护作用。
    我们使用91种循环炎症相关蛋白(N=14,824)的GWAS汇总结果进行了孟德尔随机化(MR)分析,以评估其对严重COVID-19的因果影响。COVID-19表型包括住院(N=2,095,324)和关键COVID-19(N=1,086,211)。此外,进行了敏感性分析,以评估其稳健性和可靠性。
    我们发现七种循环炎症蛋白对严重的COVID-19具有积极的因果效应。其中,血清IL-10RB水平,FGF-19和CCL-2对住院和危重的COVID-19有积极贡献(OR:1.10~1.16),而其他4种蛋白质仅对关键的COVID-19赋予风险(OR:1.07~1.16),包括EIF4EBP1、IL-7、NTF3和LIF。同时,五种蛋白对重症COVID-19的住院和进展具有保护作用(OR:0.85~0.95),包括CXCL11、CDCP1、CCL4/MIP、IFNG,和LIFR。敏感性分析不支持大多数MR分析中存在异质性。
    我们的研究揭示了严重COVID-19的风险和保护性炎症蛋白,这可能对该疾病的治疗具有重要意义。
    The relationships between circulating inflammatory proteins and COVID-19 have been observed in previous cohorts. However, it is not unclear which circulating inflammatory proteins may boost the risk of or protect against COVID-19.
    We performed Mendelian randomization (MR) analysis using GWAS summary result of 91 circulating inflammation-related proteins (N = 14,824) to assess their causal impact on severe COVID-19. The COVID-19 phenotypes encompassed both hospitalized (N = 2,095,324) and critical COVID-19 (N = 1,086,211). Moreover, sensitivity analyses were conducted to evaluate the robustness and reliability.
    We found that seven circulating inflammatory proteins confer positive causal effects on severe COVID-19. Among them, serum levels of IL-10RB, FGF-19, and CCL-2 positively contributed to both hospitalized and critical COVID-19 conditions (OR: 1.10~1.16), while the other 4 proteins conferred risk on critical COVID-19 only (OR: 1.07~1.16), including EIF4EBP1, IL-7, NTF3, and LIF. Meanwhile, five proteins exert protective effects against hospitalization and progression to critical COVID-19 (OR: 0.85~0.95), including CXCL11, CDCP1, CCL4/MIP, IFNG, and LIFR. Sensitivity analyses did not support the presence of heterogeneity in the majority of MR analyses.
    Our study revealed risk and protective inflammatory proteins for severe COVID-19, which may have vital implications for the treatment of the 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
    干扰素-τ(IFN-τ)参与反刍动物子宫内膜容受性的建立。然而,IFN-τ建立牛子宫内膜容受性的确切机制仍然未知。干扰素调节因子1(IRF1)是由I型干扰素诱导的经典干扰素刺激基因(ISG),包括IFN-τ。白血病抑制因子受体(LIFR)是白血病抑制因子(LIF)的跨膜受体,这是调节哺乳动物胚胎植入的关键因素。本研究旨在探讨IRF1和LIFR在IFN-τ调控牛子宫内膜容受性中的作用。在体内,我们发现,与发情周期的第18天相比,在妊娠第18天牛子宫内膜腔上皮中IRF1和LIFR上调.体外,IFN-τ可以上调IRF1、LIFR、和子宫内膜容受性标志物(LIF,HOXA10,ITGAV,和ITGB3)表达式,下调E-cadherin的表达并减少牛子宫内膜上皮细胞(bEECs)的微绒毛数量。IRF1过表达对子宫内膜容受性的影响与IFN-τ相似,LIFR的干扰可以阻止这些影响,提示IRF1对子宫内膜容受性的积极影响是由LIFR介导的。双荧光素酶报告基因测定证实IRF1可以通过与其启动子结合来反式激活LIFR转录。总之,IFN-τ可诱导牛子宫内膜上皮细胞IRF1表达,IRF1通过结合LIFR启动子上调LIFR表达,有助于增强牛子宫内膜的容受性。
    Interferon-τ (IFN-τ) participates in the establishment of endometrial receptivity in ruminants. However, the precise mechanisms by which IFN-τ establishes bovine endometrial receptivity remain largely unknown. Interferon regulatory factor 1 (IRF1) is a classical interferon-stimulated gene (ISG) induced by type I interferon, including IFN-τ. Leukemia inhibitory factor receptor (LIFR) is a transmembrane receptor for leukemia inhibitory factor (LIF), which is a key factor in regulating embryo implantation in mammals. This study aimed to investigate the roles of IRF1 and LIFR in the regulation of bovine endometrial receptivity by IFN-τ. In vivo, we found IRF1 and LIFR were upregulated in the bovine endometrial luminal epithelium on Day 18 of pregnancy compared to Day 18 of the estrous cycle. In vitro, IFN-τ could upregulate IRF1, LIFR, and endometrial receptivity markers (LIF, HOXA10, ITGAV, and ITGB3) expression, downregulate E-cadherin expression and reduce the quantity of microvilli of bovine endometrial epithelial cells (bEECs). Overexpression of IRF1 had similar effects to IFN-τ on endometrial receptivity, and interference of LIFR could block these effects, suggesting the positive effects of IRF1 on endometrial receptivity were mediated by LIFR. Dual luciferase reporter assay verified that IRF1 could transactivate LIFR transcription by binding to its promoter. In conclusion, IFN-τ can induce IRF1 expression in bovine endometrial epithelial cells, and IRF1 upregulates LIFR expression by binding to LIFR promoter, contributing to the enhancement of bovine endometrial receptivity.
    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
    子宫内膜癌(ECa)是最常见的女性妇科癌症。当比较子宫内膜癌的两种组织学亚型时,II型肿瘤在生物学上更具侵袭性,并且具有比I型肿瘤更差的预后。目前II型肿瘤的治疗无效,迫切需要新的靶向治疗。LIFR及其配体,LIF,已被证明在多种实体癌的进展和治疗耐药性中起关键作用。LIF/LIFR在II型ECa进展中的作用,另一方面,是未知的。我们研究了LIF/LIFR信号在II型ECa中的作用,并测试了新型小分子LIFR抑制剂EC359的功效,对抗II型ECa。对肿瘤数据库的分析揭示了生存率降低与白血病抑制因子(LIF)表达增加之间的相关性。提示在II型ECa中改变的LIF表达与不利的总体生存率之间存在潜在的联系。从细胞活力和集落形成测定获得的结果表明,与载体对照细胞相比,II型ECaLIFR敲低细胞的生长显著降低。此外,在原代和已建立的II型ECa细胞中,用EC359对LIF/LIFR轴的药理学抑制显着降低了细胞活力,细胞长期存活,和入侵,促进细胞凋亡。此外,EC359处理减少了由LIF/LIFR驱动的途径的激活,如AKT,mTOR,和STAT3。在两种不同的患者来源的体内异种移植模型和患者来源的离体器官中,EC359治疗显著抑制了肿瘤进展。总的来说,这些结果表明LIFR抑制剂EC359可能是治疗II型ECa的新的小分子治疗剂.
    Endometrial cancer (ECa) is the most common female gynecologic cancer. When comparing the two histological subtypes of endometrial cancer, Type II tumors are biologically more aggressive and have a worse prognosis than Type I tumors. Current treatments for Type II tumors are ineffective, and new targeted therapies are urgently needed. LIFR and its ligand, LIF, have been shown to play a critical role in the progression of multiple solid cancers and therapy resistance. The role of LIF/LIFR in the progression of Type II ECa, on the other hand, is unknown. We investigated the role of LIF/LIFR signaling in Type II ECa and tested the efficacy of EC359, a novel small-molecule LIFR inhibitor, against Type II ECa. The analysis of tumor databases has uncovered a correlation between diminished survival rates and increased expression of leukemia inhibitory factor (LIF), suggesting a potential connection between altered LIF expression and unfavorable overall survival in Type II ECa. The results obtained from cell viability and colony formation assays demonstrated a significant decrease in the growth of Type II ECa LIFR knockdown cells in comparison to vector control cells. Furthermore, in both primary and established Type II ECa cells, pharmacological inhibition of the LIF/LIFR axis with EC359 markedly decreased cell viability, long-term cell survival, and invasion, and promoted apoptosis. Additionally, EC359 treatment reduced the activation of pathways driven by LIF/LIFR, such as AKT, mTOR, and STAT3. Tumor progression was markedly inhibited by EC359 treatment in two different patient-derived xenograft models in vivo and patient-derived organoids ex vivo. Collectively, these results suggest LIFR inhibitor EC359 as a possible new small-molecule therapeutics for the management of Type II ECa.
    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
    导致全基因组表观遗传改变的表观遗传疗法,可以触发不同组蛋白标记之间的局部相互作用,导致表观遗传治疗的转录结果和治疗反应的转换。然而,在具有不同致癌激活的人类癌症中,致癌途径如何与表观遗传修饰因子合作来调节组蛋白标记相互作用尚不清楚.我们在此发现Hedgehog(Hh)途径重新编程乳腺癌中的组蛋白甲基化景观,尤其是三阴性乳腺癌(TNBC)。这促进了由组蛋白脱乙酰酶(HDAC)抑制剂引起的组蛋白乙酰化,并引起组合疗法的新的治疗脆弱性。具体来说,在乳腺癌中过表达小脑锌指蛋白1(ZIC1)促进Hh激活,促进H3K27甲基化(H3K27me)向乙酰化(H3K27ac)的转换。H3K27me和H3K27ac的相互排斥关系允许它们在致癌基因基因座处的功能相互作用并转换治疗结果。使用多种体内乳腺癌模型,包括患者来源的TNBC异种移植物,我们表明,Hh信号协调的H3K27me和H3K27ac在治疗乳腺癌中发挥了联合表观遗传药物的作用。一起,这项研究揭示了Hh信号调节的组蛋白修饰在响应HDAC抑制剂中的新作用,并提出了治疗TNBC的新的表观遗传靶向治疗方案。
    Epigenetic therapies that cause genome-wide epigenetic alterations, could trigger local interplay between different histone marks, leading to a switch of transcriptional outcome and therapeutic responses of epigenetic treatment. However, in human cancers with diverse oncogenic activation, how oncogenic pathways cooperate with epigenetic modifiers to regulate the histone mark interplay is poorly understood. We herein discover that the hedgehog (Hh) pathway reprograms the histone methylation landscape in breast cancer, especially in triple-negative breast cancer (TNBC). This facilitates the histone acetylation caused by histone deacetylase (HDAC) inhibitors and gives rise to new therapeutic vulnerability of combination therapies. Specifically, overexpression of zinc finger protein of the cerebellum 1 (ZIC1) in breast cancer promotes Hh activation, facilitating the switch of H3K27 methylation (H3K27me) to acetylation (H3K27ac). The mutually exclusive relationship of H3K27me and H3K27ac allows their functional interplay at oncogenic gene locus and switches therapeutic outcomes. Using multiple in vivo breast cancer models including patient-derived TNBC xenograft, we show that Hh signaling-orchestrated H3K27me and H3K27ac interplay tailors combination epigenetic drugs in treating breast cancer. Together, this study reveals the new role of Hh signaling-regulated histone modifications interplay in responding to HDAC inhibitors and suggests new epigenetically-targeted therapeutic solutions for treating TNBC.
    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:制瘤素M(OSM)是白介素(IL)-6家族的分泌细胞因子,通过激活共同信号转导成分糖蛋白130(gp130)和OSM受体β(OSMR)或白血病抑制因子受体(LIFR)的功能受体复合物来诱导生物学效应,主要涉及慢性炎症和心血管疾病。OSM/OSMR/LIFR对心肌肥厚发展的影响和潜在机制尚不清楚。
    结果:对OSMR敲除(OSMR-KO)小鼠进行主动脉束带(AB)手术,以建立压力超负荷诱导的心肌肥厚模型。超声心动图,组织学,进行了心肌的生化和免疫学分析以及骨髓源性巨噬细胞(BMDMs)的过继转移,以进行体内研究。分离BMDM并用脂多糖(LPS)刺激用于体外研究。OSMR缺乏加重了心脏肥大,小鼠AB手术后的纤维化重塑和心脏功能障碍。机械上,OSMR的缺失激活了OSM/LIFR/STAT3信号传导,并促进了促分化的巨噬细胞表型,在重塑过程中加剧了炎症和心脏修复受损.此外,AB手术后,OSMR-KOBMDMs过继性转移至WT小鼠导致一致的肥大表型。此外,用Ad-shLIFR敲除心肌组织中的LIFR改善了OSMR缺失对表型和STAT3激活的影响。
    结论:OSMR缺乏通过调节巨噬细胞和OSM/LIFR/STAT3信号传导加重压力超负荷诱导的心肌肥大,这提供了证据表明OSMR可能是治疗病理性心脏肥大和心力衰竭的有吸引力的靶标。
    Oncostatin M (OSM) is a secreted cytokine of the interleukin (IL)-6 family that induces biological effects by activating functional receptor complexes of the common signal transducing component glycoprotein 130 (gp130) and OSM receptor β (OSMR) or leukaemia inhibitory factor receptor (LIFR), which are mainly involved in chronic inflammatory and cardiovascular diseases. The effect and underlying mechanism of OSM/OSMR/LIFR on the development of cardiac hypertrophy remains unclear.
    OSMR-knockout (OSMR-KO) mice were subjected to aortic banding (AB) surgery to establish a model of pressure overload-induced cardiac hypertrophy. Echocardiographic, histological, biochemical and immunological analyses of the myocardium and the adoptive transfer of bone marrow-derived macrophages (BMDMs) were conducted for in vivo studies. BMDMs were isolated and stimulated with lipopolysaccharide (LPS) for the in vitro study. OSMR deficiency aggravated cardiac hypertrophy, fibrotic remodelling and cardiac dysfunction after AB surgery in mice. Mechanistically, the loss of OSMR activated OSM/LIFR/STAT3 signalling and promoted a proresolving macrophage phenotype that exacerbated inflammation and impaired cardiac repair during remodelling. In addition, adoptive transfer of OSMR-KO BMDMs to WT mice after AB surgery resulted in a consistent hypertrophic phenotype. Moreover, knockdown of LIFR in myocardial tissue with Ad-shLIFR ameliorated the effects of OSMR deletion on the phenotype and STAT3 activation.
    OSMR deficiency aggravated pressure overload-induced cardiac hypertrophy by modulating macrophages and OSM/LIFR/STAT3 signalling, which provided evidence that OSMR might be an attractive target for treating pathological cardiac hypertrophy and heart failure.
    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
    白血病抑制因子(LIF)是属于IL-6家族的细胞因子,在子宫植入过程中在生殖水平上很重要。然而,关于其在卵巢水平的作用的证据很少。这项工作的目的是研究LIF/LIFRβ系统在大鼠卵巢卵泡发育和类固醇生成中的局部参与。为了进行这项研究,LIF/LIFR/GP130转录物和蛋白质水平在可育和亚可育大鼠卵巢中测量,并进行体外实验以评估STAT3的激活。然后,在体内实验中,通过渗透微型泵将LIF长期和局部施用于大鼠卵巢28天,以使我们能够评估对卵泡生成和类固醇生成的影响。通过q-PCR和WB确定LIF及其受体存在于可育和亚可育卵巢中,LIF在发情周期中变化,在发情期和中/发情期阶段更高。除此之外,发现LIF可以激活STAT3通路并引起pSTAT3的形成。还观察到LIF减少了窦前和窦卵泡的数量和大小,而不改变闭锁的窦卵泡的数量。可以增加黄体的数量,孕酮水平显着增加。因此,可以推断LIF在体内对卵泡发生具有重要作用,排卵和类固醇生成,特别是黄体酮的合成。
    Leukaemia inhibitory factor (LIF) is a cytokine belonging to the interleukin-6 family that is important at the reproductive level in the uterine implantation process. However, there is very little evidence regarding its effect at the ovarian level. The aim of this work was to study the local involvement of the LIF/LIFRβ system in follicular development and steroidogenesis in rat ovaries. To carry out this research, LIF/LIFR/GP130 transcript and protein levels were measured in fertile and sub-fertile rat ovaries, and in vitro experiments were performed to assess STAT3 activation. Then, in in vivo experiments, LIF was administered chronically and locally for 28 days to the ovaries of rats by means of an osmotic minipump to enable us to evaluate the effect on folliculogenesis and steroidogenesis. It was determined by quantitative polymerase chain reaction and western blot that LIF and its receptors are present in fertile and sub-fertile ovaries and that LIF varies during the oestrous cycle, being higher during the oestrus and meta/dioestrus stages. In addition to this, it was found that LIF can activate STAT3 pathways and cause pSTAT3 formation. It was also observed that LIF decreases the number and size of preantral and antral follicles without altering the number of atretic antral follicles and can increase the number of corpora lutea, with a notable increase in the levels of progesterone (P4). It is therefore possible to infer that LIF exerts an important effect in vivo on folliculogenesis, ovulation and steroidogenesis, specifically the synthesis of P4.
    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
    白血病抑制因子(LIF),是属于IL-6家族的细胞因子,其过度表达与癌症患者的不良预后相关,包括胰腺导管腺癌(PDAC)。LIF信号传导通过其与由LIFR受体和Gp130形成的异二聚体LIF受体(LIFR)复合物的结合来介导,导致JAK1/STAT3激活。胆汁酸是类固醇,调节膜和核受体的表达/活性,包括法尼醇-X-受体(FXR)和G蛋白胆汁酸激活受体(GPBAR1)。
    本文中,我们研究了FXR和GPBAR1的配体是否调节PDAC细胞中的LIF/LIFR途径以及这些受体是否在人肿瘤组织中表达。
    一组PDCA患者的转录组分析显示,与配对的非肿瘤组织相比,肿瘤组织中LIF和LIFR的表达增加。通过体外测定,我们发现初级和次级胆汁酸对LIF/LIFR信号传导都具有弱的拮抗作用。相比之下,BAR502一种非胆汁酸甾体双FXR和GPBAR1配体,有效抑制LIF与LIFR的结合,IC50为3.8µM。
    BAR502以FXR和GPBAR1独立的方式反转LIF诱导的模式,提示BAR502在LIFR过表达PDAC治疗中的潜在作用。
    UNASSIGNED: The leukemia inhibitory factor (LIF), is a cytokine belonging to IL-6 family, whose overexpression correlate with poor prognosis in cancer patients, including pancreatic ductal adenocarcinoma (PDAC). LIF signaling is mediate by its binding to the heterodimeric LIF receptor (LIFR) complex formed by the LIFR receptor and Gp130, leading to JAK1/STAT3 activation. Bile acids are steroid that modulates the expression/activity of membrane and nuclear receptors, including the Farnesoid-X-Receptor (FXR) and G Protein Bile Acid Activated Receptor (GPBAR1).
    UNASSIGNED: Herein we have investigated whether ligands to FXR and GPBAR1 modulate LIF/LIFR pathway in PDAC cells and whether these receptors are expressed in human neoplastic tissues.
    UNASSIGNED: The transcriptome analysis of a cohort of PDCA patients revealed that expression of LIF and LIFR is increased in the neoplastic tissue in comparison to paired non-neoplastic tissues. By in vitro assay we found that both primary and secondary bile acids exert a weak antagonistic effect on LIF/LIFR signaling. In contrast, BAR502 a non-bile acid steroidal dual FXR and GPBAR1 ligand, potently inhibits binding of LIF to LIFR with an IC50 of 3.8 µM.
    UNASSIGNED: BAR502 reverses the pattern LIF-induced in a FXR and GPBAR1 independent manner, suggesting a potential role for BAR502 in the treatment of LIFR overexpressing-PDAC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号