Zinc Finger Protein GLI1

锌指蛋白 GLI1
  • 文章类型: Journal Article
    黄韧带(LF)肥大是腰椎管狭窄(LSCS)的重要因素。lncRNA在器官纤维化中起着至关重要的作用,但其在LF纤维化中的作用尚不清楚。我们之前的研究结果表明,Hedgehog-Gli1信号传导是导致LF肥大的关键驱动因素。通过RIP实验,我们小组发现lnc-RMRP与Gli1物理相关,并在Gli1激活的LF细胞中表现出富集。组织学研究显示,肥厚性LF中RMRP的表达升高。体外实验进一步证实RMRP促进Gli1SUMO修饰和核转移。机械上,RMRP诱导GSDMD介导的焦亡,促炎激活,和胶原蛋白通过Hedgehog途径表达。值得注意的是,机械应力诱导的兔LF肥大表现出类似的人LF纤维化病理变化,并显示胶原蛋白和α-SMA水平升高。RMRP敲低导致纤维化和焦亡相关蛋白表达降低,最终改善纤维化。以上数据得出结论,RMRP在通过Gli1SUMO化调节GSDMD介导的LF细胞焦亡中起关键作用,因此表明靶向RMRP可以作为LF肥大和纤维化的潜在和有效的治疗策略.
    Hypertrophy of ligamentum flavum (LF) is a significant contributing factor to lumbar spinal canal stenosis (LSCS). lncRNA plays a vital role in organ fibrosis, but its role in LF fibrosis remains unclear. Our previous findings have demonstrated that Hedgehog-Gli1 signaling is a critical driver leading to LF hypertrophy. Through the RIP experiment, our group found lnc-RMRP was physically associated with Gli1 and exhibited enrichment in Gli1-activated LF cells. Histological studies revealed elevated expression of RMRP in hypertrophic LF. In vitro experiments further confirmed that RMRP promoted Gli1 SUMO modification and nucleus transfer. Mechanistically, RMRP induced GSDMD-mediated pyroptosis, proinflammatory activation, and collagen expression through the Hedgehog pathway. Notably, the mechanical stress-induced hypertrophy of LF in rabbit exhibited analogous pathological changes of LF fibrosis occurred in human and showed enhanced levels of collagen and α-SMA. Knockdown of RMRP resulted in the decreased expression of fibrosis and pyroptosis-related proteins, ultimately ameliorating fibrosis. The above data concluded that RMRP exerts a crucial role in regulating GSDMD-mediated pyroptosis of LF cells via Gli1 SUMOylation, thus indicating that targeting RMRP could serve as a potential and effective therapeutic strategy for LF hypertrophy and fibrosis.
    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
    套细胞淋巴瘤(MCL)是一种罕见且侵袭性的非霍奇金淋巴瘤。其治疗的挑战包括复发,耐药性,生存期短。Hedgehog/GLI1(Hh/GLI1)和Wnt/β-catenin通路在癌细胞增殖中至关重要,生存,和抗药性,使它们成为抗癌研究的重要目标。本研究旨在评估两种途径联合抑制剂对MCL的有效性,并研究潜在的分子机制。在MCL中观察到来自Hh/GLI1和Wnt/β-catenin途径的关键蛋白的共表达。用GLI1抑制剂GANT61靶向Hh/GLI1途径和用CBP/β-catenin转录抑制剂ICG-001靶向Wnt/β-catenin途径,双靶向治疗被证明协同抑制MCL细胞的活性。这种方法促进MCL细胞凋亡,诱导G0/G1期阻滞,降低了S期细胞的百分比,并增强MCL细胞对药物阿霉素和依鲁替尼的敏感性。GANT61和ICG-001均下调GLI1和β-连环蛋白,同时上调GSK-3β表达。Hh/GLI1和Wnt/β-catenin途径之间的相互作用是由GANT61依赖性Hh/GLI1抑制介导的。此外,GLI1敲除联合ICG-001协同诱导MCL细胞凋亡并增加对阿霉素和依鲁替尼的药物敏感性。GANT61减弱了MCL细胞中β-catenin的过表达并降低了GSK-3β的抑制作用。总的来说,Hh/GLI1和Wnt/β-catenin通路的联合靶向在抑制增殖方面更有效,诱导G0/G1周期延迟,促进细胞凋亡,与单一治疗相比,MCL细胞的药物敏感性增加。这些发现强调了联合疗法治疗MCL患者的潜力。
    Mantle cell lymphoma (MCL) is a rare and aggressive form of non-Hodgkin lymphoma. Challenges in its treatment include relapse, drug resistance, and a short survival period. The Hedgehog/GLI1 (Hh/GLI1) and Wnt/β-catenin pathways are crucial in cancer cell proliferation, survival, and drug resistance, making them significant targets for anticancer research. This study aimed to assess the effectiveness of combining inhibitors for both pathways against MCL and investigate the underlying molecular mechanisms. The co-expression of key proteins from the Hh/GLI1 and Wnt/β-catenin pathways was observed in MCL. Targeting the Hh/GLI1 pathway with the GLI1 inhibitor GANT61 and the Wnt/β-catenin pathway with the CBP/β-catenin transcription inhibitor ICG-001, dual-target therapy was demonstrated to synergistically suppressed the activity of MCL cells. This approach promoted MCL cell apoptosis, induced G0/G1 phase blockade, decreased the percentage of S-phase cells, and enhanced the sensitivity of MCL cells to the drugs adriamycin and ibrutinib. Both GANT61 and ICG-001 downregulated GLI1 and β-catenin while upregulating GSK-3β expression. The interaction between Hh/GLI1 and Wnt/β-catenin pathways was mediated by GANT61-dependent Hh/GLI1 inhibition. Moreover, GLI1 knockdown combined with ICG-001 synergistically induced apoptosis and increased drug sensitivity of MCL cells to doxorubicin and ibrutinib. GANT61 attenuated the overexpression of β-catenin and decreased the inhibition of GSK-3β in MCL cells. Overall, the combined targeting of both the Hh/GLI1 and Wnt/β-catenin pathways was more effective in suppressing proliferation, inducing G0/G1 cycle retardation, promoting apoptosis, and increasing drug sensitivity of MCL cells than mono treatments. These findings emphasize the potential of combinatorial therapy for treating MCL patients.
    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
    编码NUAK家族激酶1(NUAK1)的基因经常被扩增,其表达上调,在各种癌症中激活致癌信号。然而,对其在胃癌(GC)中的作用知之甚少。我们调查了NUAK1、刺猬信号、和GC的肿瘤发生。NUAK1过表达在本地和公共GC队列中得到验证。患者来源的异种移植和转基因小鼠模型证明NUAK1消耗或抑制显著改善胃肿瘤发生。NUAK1通过激活STAT5介导的转录和稳定GLI1蛋白上调GLI1表达。NUAK1消耗或抑制损害癌细胞扩增,肿瘤形成,和体外和体内模型中的化疗抗性。临床病理分析证实,NUAK1表达上调与人GC预后不良和化疗耐药相关。我们的发现表明信号轴NUAK1/STAT5/GLI1促进癌细胞扩增和肿瘤发生,并表明NUAK1是GC中一个有吸引力的治疗靶标和预后因素。
    The gene encoding the NUAK family kinase 1 (NUAK1) is frequently amplified and its expression is upregulated, activating oncogenic signaling in various cancers. However, little is known about its role in gastric cancer (GC). We investigate the mechanistic links among NUAK1, Hedgehog signaling, and tumorigenesis in GC. NUAK1 overexpression is validated in local and public GC cohorts. Patient-derived xenograft and transgenic mouse models demonstrate that NUAK1 depletion or inhibition dramatically ameliorates gastric tumorigenesis. NUAK1 upregulates GLI1 expression by activating STAT5-mediated transcription and stabilizing GLI1 protein. NUAK1 depletion or inhibition impairs cancer cell expansion, tumor formation, and chemotherapy resistance in in vitro and in vivo models. Clinicopathological analysis confirms that upregulated NUAK1 expression correlates with poor prognosis and chemotherapy resistance in human GC. Our findings demonstrate that the signaling axis NUAK1/STAT5/GLI1 promotes cancer cell expansion and tumorigenesis and indicate that NUAK1 is an attractive therapeutic target and prognostic factor in GC.
    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:跨缝线牵张成骨(TSDO)涉及对面部缝线施加牵张力以刺激成骨。颅骨缝合线中的Gli1细胞在骨骼生长中起重要作用。然而,面部缝合线中的Gli1+细胞在牵张力下是否分化成骨未知。
    方法:使用4周龄Gli1ER/Td和C57BL/6小鼠建立TSDO模型,以探讨吻合齿吻合缝的成骨作用。采用Gli1+细胞谱系示踪模型观察Gli1+细胞分布,探讨Gli1+细胞在面骨重建中的作用。
    结果:牵张力促进TSDO期间的骨重建。荧光和双光子扫描图像揭示了Gli1细胞的分布。在分心的力量下,Gli1谱系细胞显著增殖并与Runx2+细胞共定位。Hedgehog信号在Gli1+细胞中上调。Hedgehog信号的抑制抑制由牵张力诱导的Gli1+细胞的增殖和成骨。随后,鉴定了Gli1+细胞的干细胞特性。细胞拉伸实验验证了机械力通过Hh信号促进Gli1+细胞的成骨分化。此外,免疫荧光染色和RT-qPCR实验表明,Gli1细胞中的初级纤毛表现出不依赖Hedgehog的机械敏感性,这是机械力诱导的成骨分化所必需的。
    结论:我们的研究表明,Gli1+细胞的初级纤毛感觉到机械刺激,介导Hedgehog信号激活,促进Gli1+细胞在腋窝缝合中的成骨分化。
    BACKGROUND: Trans-sutural distraction osteogenesis (TSDO) involves the application of distraction force to facial sutures to stimulate osteogenesis. Gli1+ cells in the cranial sutures play an important role in bone growth. However, whether Gli1+ cells in facial sutures differentiate into bone under distraction force is unknown.
    METHODS: 4-week-old Gli1ER/Td and C57BL/6 mice were used to establish a TSDO model to explore osteogenesis of zygomaticomaxillary sutures. A Gli1+ cell lineage tracing model was used to observe the distribution of Gli1+ cells and explore the role of Gli1+ cells in facial bone remodeling.
    RESULTS: Distraction force promoted bone remodeling during TSDO. Fluorescence and two-photon scanning images revealed the distribution of Gli1+ cells. Under distraction force, Gli1-lineage cells proliferated significantly and co-localized with Runx2+ cells. Hedgehog signaling was upregulated in Gli1+ cells. Inhibition of Hedgehog signaling suppresses the proliferation and osteogenesis of Gli1+ cells induced by distraction force. Subsequently, the stem cell characteristics of Gli1+ cells were identified. Cell-stretching experiments verified that mechanical force promoted the osteogenic differentiation of Gli1+ cells through Hh signaling. Furthermore, immunofluorescence staining and RT-qPCR experiments demonstrated that the primary cilia in Gli1+ cells exhibit Hedgehog-independent mechanosensitivity, which was required for the osteogenic differentiation induced by mechanical force.
    CONCLUSIONS: Our study indicates that the primary cilia of Gli1+ cells sense mechanical stimuli, mediate Hedgehog signaling activation, and promote the osteogenic differentiation of Gli1+ cells in zygomaticomaxillary sutures.
    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
    Hedgehog(Hh)信号通路参与T细胞的分化和发育,在T细胞发育的不同阶段起着重要的调节作用。我们先前的研究表明,产前暴露于葡萄球菌肠毒素B(SEB)会改变后代胸腺中T细胞亚群的百分比。然而,目前尚不清楚产前SEB暴露是否会影响胸腺T细胞中的Hh信号通路.在本研究中,妊娠第16天的怀孕大鼠静脉注射15μgSEB一次,对新生和成年后代大鼠的胸腺进行无菌采集,以研究SEB对Hh信号通路的影响。首先发现产前SEB暴露明显导致新生和成年子代大鼠胸腺组织中Hh信号通路Shh和Dhh配体表达增加,但显著降低了膜受体Ptch1和Smo的表达水平,转录因子Gli1,以及CyclinD1,C-myc,和N-myc在胸腺T细胞Hh信号通路中的作用。这些数据表明,产前SEB暴露抑制了新生儿后代胸腺T淋巴细胞中的Hh信号通路,这种效应可以通过印记效应在成年后代中保持。
    The Hedgehog (Hh) signaling pathway is involved in T cell differentiation and development and plays a major regulatory part in different stages of T cell development. A previous study by us suggested that prenatal exposure to staphylococcal enterotoxin B (SEB) changed the percentages of T cell subpopulation in the offspring thymus. However, it is unclear whether prenatal SEB exposure impacts the Hh signaling pathway in thymic T cells. In the present study, pregnant rats at gestational day 16 were intravenously injected once with 15 μg SEB, and the thymi of both neonatal and adult offspring rats were aseptically acquired to scrutinize the effects of SEB on the Hh signaling pathway. It firstly found that prenatal SEB exposure clearly caused the increased expression of Shh and Dhh ligands of the Hh signaling pathway in thymus tissue of both neonatal and adult offspring rats, but significantly decreased the expression levels of membrane receptors of Ptch1 and Smo, transcription factor Gli1, as well as target genes of CyclinD1, C-myc, and N-myc in Hh signaling pathway of thymic T cells. These data suggest that prenatal SEB exposure inhibits the Hh signaling pathway in thymic T lymphocytes of the neonatal offspring, and this effect can be maintained in adult offspring via the imprinting effect.
    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: Case Reports
    背景技术软组织肿瘤具有多种亚型,其中肉瘤表现出高恶性潜能和不良预后。最初在具有t(7;12)易位的肌周细胞瘤中发现了具有GLI1改变的恶性上皮样肿瘤。然而,最近的研究表明,它是一种独特的肿瘤类型,其特征是椭圆形或圆形上皮样细胞的多个结节状分布,具有丰富的毛细血管网络和缺乏特异性的免疫表型。世界上只有少数病例报告,最佳治疗仍在探索中。病例报告我们报告了一例31岁的患者,该患者表现为严重贫血和十二指肠大的软组织肿块。患者接受手术切除,切缘阴性,15个淋巴结中没有一个肿瘤检测呈阳性。术后病理和FISH测试进一步证实存在GLI1破坏以及S-100和SMA阴性。基因检测显示ACTB-GLI1融合。手术后没有提供特定的药物。在23个月的随访期间未发现肿瘤复发。目前患者的生活质量令人满意。结论以GLI1基因重排为特征的软组织肉瘤具有相对较低的侵袭性和转移性,固体物质即使在生长时也会最小程度地传播。患者可以从手术切除中受益,导致相对较长的无瘤生存期。
    BACKGROUND Soft tissue tumors have various subtypes, among which sarcomas exhibit high malignant potential and poor prognosis. Malignant epithelioid tumor with GLI1 alterations was originally found in myopericytoma with t(7;12) translocation. However, recent studies indicated that it is a distinct tumor type characterized by multiple nodular distributions of oval or round epithelioid cells with a rich capillary network and a lack of specific immunophenotype. There are only a few cases reported worldwide and the optimal treatment is still being explored. CASE REPORT We report the case of a 31-year-old patient who presented with severe anemia and a large soft tissue mass in the duodenum. The patient underwent surgical resection with a negative margin, and none of the 15 lymph nodes tested positive for the tumor. Postoperative pathology and FISH testing further confirmed the presence of GLI1 disruption and S-100 and SMA negativity. Genetic testing revealed the ACTB-GLI1 fusion. No specific medication was offered after the surgery. No tumor recurrence was found during the 23-month follow-up period. The patient\'s quality of life is currently satisfactory. CONCLUSIONS Soft tissue sarcomas characterized by GLI1 gene rearrangement have a relatively less aggressive and metastatic nature, with the solid mass spreading minimally even as it grows. Patients can benefit from surgical resection, resulting in a relatively long period of tumor-free survival.
    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:目前的证据表明,由于各种不良反应,没有完全有效的子宫内膜异位症(EMS)方法而没有创伤。可靠的证据表明,抑制铁死亡是EMS的潜在策略。我们充分证实内源性蛋白PDZ和LIM结构域3(PDLIM3)在EMS中的表达显著增加。
    方法:PDLIM3敲低降低了原发性异位子宫内膜基质细胞(EESCs)的活力和迁移,和升高的铁凋亡信号指标,包括Fe2+,丙二醛(MDA),EESCs中的活性氧(ROS)。
    结果:机制研究表明,抑制PDLIM3加速了神经胶质瘤相关癌基因-1(Gli1)的降解,并进一步使Hedgehog信号失活。Gli1抑制剂,GANT61,消除了PDLIM3缺失对EESC生长的影响,迁移,和铁中毒。体内实验表明,PDLIM3的减少抑制了子宫内膜病变的生长。同样,抑制PDLIM3可促进子宫内膜病变中的铁凋亡并减弱Hedgehog信号传导。
    结论:总的来说,PDLIM3的沉默通过诱导Gli1降解和阻断Hedgehog信号传导促进EMS中的铁凋亡。它可能为将来开发EMS的治疗剂提供替代策略。
    OBJECTIVE: Current evidence suggests that there is no completely effective method for endometriosis (EMS) without trauma due to diverse adverse effects. Reliable evidence illustrates that inhibiting ferroptosis is a potential strategy for EMS. We sufficiently verified that the expression of endogenous protein PDZ and LIM domain 3 (PDLIM3) was significantly increased in EMS.
    METHODS: PDLIM3 knockdown reduced primary ectopic endometrial stromal cells\' (EESCs) viability and migration, and elevated ferroptosis signaling indicators including Fe2+, malondialdehyde (MDA), and reactive oxygen species (ROS) in EESCs.
    RESULTS: Mechanistic studies revealed that inhibition of PDLIM3 accelerated glioma-associated oncogene-1 (Gli1) degradation and further deactivated Hedgehog signaling. Gli1 inhibitor, GANT61, abrogated the impact of PDLIM3 deletion on EESC growth, migration, and ferroptosis. In vivo experiments suggested that PDLIM3 reduction repressed the growth of endometrial lesions. Likewise, repression of PDLIM3 promoted ferroptosis and attenuated Hedgehog signaling in endometrial lesions.
    CONCLUSIONS: Collectively, silencing of PDLIM3 facilitates ferroptosis in EMS by inducing Gli1 degradation and blocking Hedgehog signaling. It may provide an alternative strategy for developing therapeutic agents of EMS in the future.
    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
    N6-甲基腺苷(m6A)修饰,由甲基转移酶样3(METTL3)催化的真核信使RNA修饰,在干细胞命运决定中起着举足轻重的作用。颅骨的发育和维持是由颅骨缝合线协调的。组织蛋白酶K(CTSK)阳性的颅骨干细胞(CSC)有助于小鼠颅骨骨化。然而,在颅骨发育过程中,m6A修饰在调节Ctsk谱系细胞中的作用仍然难以捉摸。这里,我们显示METTL3与颅骨非破骨细胞Ctsk谱系细胞共定位,也与GLI1表达相关。在新生儿发育过程中,Ctsk谱系细胞中Mettl3的消耗延迟了缝合线的形成并减少了矿化。在成年期间维持,Ctsk+谱系细胞中Mettl3的丢失损害了颅骨形成,其特点是骨骼孔隙率增加,增强的骨髓腔,细胞轮廓不发达的骨细胞数量减少。甲基化RNA免疫沉淀测序和RNA测序数据的分析表明METTL3的缺失降低了Hedgehog(Hh)信号通路。通过交叉Sufufl/+等位基因或通过局部施用SAG21来恢复Hh信号通路部分地挽救了该异常。我们的数据表明,METTL3通过调节Hh信号通路调节支持颅骨骨形成的Ctsk+谱系细胞,为颅骨穹窿骨病的临床治疗提供新的见解。
    N6-methyladenosine (m6A) modification, a eukaryotic messenger RNA modification catalyzed by methyltransferase-like 3 (METTL3), plays a pivotal role in stem cell fate determination. Calvarial bone development and maintenance are orchestrated by the cranial sutures. Cathepsin K (CTSK)-positive calvarial stem cells (CSCs) contribute to mice calvarial ossification. However, the role of m6A modification in regulating Ctsk+ lineage cells during calvarial development remains elusive. Here, we showed that METTL3 was colocalized with cranial nonosteoclastic Ctsk+ lineage cells, which were also associated with GLI1 expression. During neonatal development, depletion of Mettl3 in the Ctsk+ lineage cells delayed suture formation and decreased mineralization. During adulthood maintenance, loss of Mettl3 in the Ctsk+ lineage cells impaired calvarial bone formation, which was featured by the increased bone porosity, enhanced bone marrow cavity, and decreased number of osteocytes with the less-developed cellular outline. The analysis of methylated RNA immunoprecipitation sequencing and RNA sequencing data indicated that loss of METTL3 reduced Hedgehog (Hh) signaling pathway. Restoration of Hh signaling pathway by crossing Sufufl/+ alleles or by local administration of SAG21 partially rescued the abnormity. Our data indicate that METTL3 modulates Ctsk+ lineage cells supporting calvarial bone formation by regulating the Hh signaling pathway, providing new insights for clinical treatment of skull vault osseous 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: Letter
    暂无摘要。
    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: Case Reports
    暂无摘要。
    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