GLI3

Gli3
  • 文章类型: Journal Article
    背景:多指畸形是一种普遍的先天性畸形,在中国每1000例活产中有2.14例。GLI家族锌指3(GLI3)是多指的经典致病基因,并作为刺猬信号通路中的关键转录因子,调节四肢前后轴的发育。
    方法:选取湖南省3家多指患者,中国。通过全外显子组测序(WES)和Sanger测序鉴定致病变体。
    结果:在三个不相关的家族中鉴定出GLI3的三个变异体,包括一个新的缺失变体(c.1372del,p.Thr458GlnfsTer44),一个新的插入-删除(indel)变体(c.1967_1968delinsAA,p.Ser656Ter),和无义变体(c.2374C>T,p.Arg792Ter)。这些变体仅存在于患者中,而不存在于健康个体中。
    结论:我们在多指患者中发现了三种致病性GLI3变异,拓宽GLI3的遗传谱,并为多指的遗传咨询和诊断做出重要贡献。
    BACKGROUND: Polydactyly is a prevalent congenital anomaly with an incidence of 2.14 per 1000 live births in China. GLI family zinc finger 3 (GLI3) is a classical causative gene of polydactyly, and serves as a pivotal transcription factor in the hedgehog signaling pathway, regulating the development of the anterior-posterior axis in limbs.
    METHODS: Three pedigrees of polydactyly patients were enrolled from Hunan Province, China. Pathogenic variants were identified by whole-exome sequencing (WES) and Sanger sequencing.
    RESULTS: Three variants in GLI3 were identified in three unrelated families, including a novel deletion variant (c.1372del, p.Thr458GlnfsTer44), a novel insertion-deletion (indel) variant (c.1967_1968delinsAA, p.Ser656Ter), and a nonsense variant (c.2374 C > T, p.Arg792Ter). These variants were present exclusively in patients but not in healthy individuals.
    CONCLUSIONS: We identified three pathogenic GLI3 variants in polydactyly patients, broadening the genetic spectrum of GLI3 and contributing significantly to genetic counseling and diagnosis for polydactyly.
    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
    哮喘是儿童气道中最常见的慢性炎症性疾病。哮喘最普遍的表型是嗜酸性粒细胞性哮喘,这是由Th2免疫反应驱动的,可以通过吸入皮质类固醇治疗有效地管理。然而,有Th17免疫应答的哮喘表型对皮质类固醇治疗不敏感,表现出更严重的表型.这种对皮质类固醇不敏感的哮喘的治疗目前尚不成熟,需要进一步关注。本研究旨在阐明Hedgehog信号通路在哮喘Th17细胞分化中的调控作用。研究表明,Smo和Gli3都是Hedgehog信号通路的关键组成部分,在体外Th17极化和体内Th17显性哮喘模型中上调。发现用小分子抑制剂抑制Smo或基因敲除Gli3可以抑制Th17极化。发现Smo在Th1,Th2,Th17和Treg极化中增加,而Gli3在Th17极化中特异性增加。ChIP-qPCR分析表明,Gli3可以直接与T细胞中的IL-6相互作用,诱导STAT3磷酸化并促进Th17细胞分化。此外,该研究表明哮喘患儿Gli3表达升高与IL-17A和IL-6表达之间存在相关性.总之,研究表明,Hedgehog信号通路在哮喘的发病机制中起着重要作用,因为它通过IL-6/STAT3信号调节Th17细胞的分化。这可能为Th17细胞驱动的皮质类固醇不敏感哮喘提供潜在的治疗靶标。
    Asthma is the most prevalent chronic inflammatory disease of the airways in children. The most prevalent phenotype of asthma is eosinophilic asthma, which is driven by a Th2 immune response and can be effectively managed by inhaled corticosteroid therapy. However, there are phenotypes of asthma with Th17 immune response that are insensitive to corticosteroid therapy and manifest a more severe phenotype. The treatment of this corticosteroid-insensitive asthma is currently immature and requires further attention. The objective of this study is to elucidate the regulation of the Hedgehog signaling pathway in Th17 cell differentiation in asthma. The study demonstrated that both Smo and Gli3, key components of the Hedgehog signaling pathway, were upregulated in Th17 polarization in vitro and in a Th17-dominant asthma model in vivo. Inhibiting Smo with a small molecule inhibitor or genetically knocking down Gli3 was found to suppress Th17 polarization. Smo was found to increase in Th1, Th2, Th17 and Treg polarization, while Gli3 specifically increased in Th17 polarization. ChIP-qPCR analyses indicated that Gli3 can directly interact with IL-6 in T cells, inducing STAT3 phosphorylation and promoting Th17 cell differentiation. Furthermore, the study demonstrated a correlation between elevated Gli3 expression and IL-17A and IL-6 expression in children with asthma. In conclusion, the study demonstrated that the Hedgehog signaling pathway plays an important role in the pathogenesis of asthma, as it regulates the differentiation of Th17 cells through the IL-6/STAT3 signaling. This may provide a potential therapeutic target for corticosteroid-insensitive asthma driven by Th17 cells.
    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
    乳腺癌仍然是全球癌症死亡的第二大原因,因此突出了为这种疾病寻找卓越治疗策略的迫切需要。在当前的癌症治疗时代,个性化的医学正在引起很多关注,因为这种治疗方式更具选择性,从而最大限度地减少有害的副作用。个性化医学取决于了解初始肿瘤的潜在遗传景观。在我们的研究中,我们集中研究了一个在Mediator亚基12(MED12)中具有遗传改变的特定乳腺癌亚群.我们的结果表明,MED12的缺失通过涉及GLI3依赖性SHH信号激活的机制导致乳腺癌细胞的细胞增殖和集落形成增强,这是对乳房发育和体内平衡至关重要的途径。为了找到针对这一部分乳腺癌的个性化治疗选择,我们采用天然化合物筛选策略,共发现10种选择性靶向MED12敲低乳腺癌细胞的化合物.我们的结果表明,这十个化合物中的两个,solasonineandalisolB23-acetate,阻断GLI3依赖性SHH信号,导致细胞增殖和集落形成能力增强的逆转。因此,我们的研究结果为MED12改变的乳腺癌患者提供了一种新的个性化治疗策略。
    Breast cancer remains to be the second leading cause of cancer deaths worldwide thereby highlighting the critical need to find superior treatment strategies for this disease. In the current era of cancer treatment, personalized medicine is garnering much attention as this type of treatment is more selective thereby minimizing harmful side effects. Personalized medicine is dependent upon knowing the underlying genetic landscape of the initial tumor. In our study, we focused our efforts on a specific subset of breast cancer that harbors genetic alterations in the Mediator subunit 12 (MED12). Our results show that loss of MED12 leads to enhanced cellular proliferation and colony formation of breast cancer cells through a mechanism that involves activation of GLI3-dependent SHH signaling, a pathway that is central to breast development and homeostasis. To find a personalized treatment option for this subset of breast cancer, we employed a natural compound screening strategy which uncovered a total of ten compounds that selectively target MED12 knockdown breast cancer cells. Our results show that two of these ten compounds, solasonine and alisol B23-acetate, block GLI3-dependent SHH signaling which leads to a reversal of enhanced cellular proliferation and colony formation ability. Thus, our findings provide promising insight into a novel personalized treatment strategy for patients suffering from MED12-altered breast cancer.
    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:多指,尤其是食指,仍然是一个有趣的异常,没有特定的基因或基因座与该表型明确相关。在这项研究中,我们对一个显示食指多指的三代家庭进行了调查。
    方法:对患者进行外显子组测序,通过过滤来识别潜在的因果变异。通过Sanger测序对获得的变体进行验证,包括所有家庭成员。
    结果:外显子组分析在先证者和所有受影响的家族成员中的GLI3蛋白的锌指DNA结合域发现了一个新的杂合错义变体(c.1482A>T;p.Gln494His)。值得注意的是,该变异体在谱系内未受影响的个体中不存在,强调其与多指表型的关联。计算分析表明,GLI3p.Gln494His会影响跨物种高度保守的残基。
    结论:GLI3锌指DNA结合区是Sonichedgehog信号通路的重要组成部分,通过调节靶基因表达来协调胚胎发育的关键方面。这项新发现不仅为胚胎发育过程中控制多指的分子途径提供了有价值的见解,而且还具有在临床环境中增强这种疾病的诊断和筛查能力的潜力。
    BACKGROUND: Polydactyly, particularly of the index finger, remains an intriguing anomaly for which no specific gene or locus has been definitively linked to this phenotype. In this study, we conducted an investigation of a three-generation family displaying index finger polydactyly.
    METHODS: Exome sequencing was conducted on the patient, with a filtration to identify potential causal variation. Validation of the obtained variant was conducted by Sanger sequencing, encompassing all family members.
    RESULTS: Exome analysis uncovered a novel heterozygous missense variant (c.1482A>T; p.Gln494His) at the zinc finger DNA-binding domain of the GLI3 protein within the proband and all affected family members. Remarkably, the variant was absent in unaffected individuals within the pedigree, underscoring its association with the polydactyly phenotype. Computational analyses revealed that GLI3 p.Gln494His impacts a residue that is highly conserved across species.
    CONCLUSIONS: The GLI3 zinc finger DNA-binding region is an essential part of the Sonic hedgehog signaling pathway, orchestrating crucial aspects of embryonic development through the regulation of target gene expression. This novel finding not only contributes valuable insights into the molecular pathways governing polydactyly during embryonic development but also has the potential to enhance diagnostic and screening capabilities for this condition in clinical settings.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    GLI3基因突变可导致各种形式的多指,如Greig头孢多症综合征(GCPS,MIM:#175700),Pallister-Hall综合征(PHS,MIM:#146510),和孤立的多指(IPD,MIM:#174200,#174700)。关于IPD相关GLI3突变的报道很少。在这项研究中,在一个患有IPD的中国家庭中发现了一个新的GLI3突变。
    我们报告了一个有六个成员受IPD影响的家庭。家庭成员表现出几种特殊的表型,包括性别差异,手指关节发育异常,和不同的多指类型。我们在GLI3基因中发现了一个新的移码变体(NM_000168.6:c.1820_1821del,NP_000159.3:p.Tyr607Cysfs*9)通过全外显子组测序。进一步分析表明,该突变是该家族多指的原因。
    在我们的研究中发现这种新颖的移码变体进一步巩固了IPD与GLI3之间的关系,并扩展了先前建立的GLI3突变和相关表型的谱。
    UNASSIGNED: GLI3 gene mutations can result in various forms of polysyndactyly, such as Greig cephalopolysyndactyly syndrome (GCPS, MIM: #175700), Pallister-Hall syndrome (PHS, MIM: #146510), and isolated polydactyly (IPD, MIM: #174200, #174700). Reports on IPD-associated GLI3 mutations are rare. In this study, a novel GLI3 mutation was identified in a Chinese family with IPD.
    UNASSIGNED: We report a family with six members affected by IPD. The family members demonstrated several special phenotypes, including sex differences, abnormal finger joint development, and different polydactyly types. We identified a novel frameshift variant in the GLI3 gene (NM_000168.6: c.1820_1821del, NP_000159.3: p.Tyr607Cysfs*9) by whole-exome sequencing. Further analysis suggested that this mutation was the cause of polydactyly in this family.
    UNASSIGNED: The discovery of this novel frameshift variant in our study further solidifies the relationship between IPD and GLI3 and expands the previously established spectrum of GLI3 mutations and associated phenotypes.
    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
    前列腺癌(PCa)是全球男性人群中最普遍的死亡原因。G蛋白偶联雌激素受体(GPER)在PCa的开发中具有重要意义。Hedgehog(Hh)通路激活与侵袭性有关,转移,PCa患者复发。迄今为止,没有研究评估PCa中GPER与Hh通路沿不同组等级的串扰.我们使用免疫组织化学对来自不同PCa预后等级的患者的石蜡包埋组织进行了分析。评估了GPER和胶质瘤相关癌基因同源物(GLI)转录因子在PCa肿瘤实质和间质中的表达及其相关性。我们的结果表明,GPER在细胞核中高度表达,并随着高等级组的增加而增加。此外,GPER的表达与PCa组织中不同级别组的pGLI3核表达相关;然而,受体是否诱导GLI转录因子的激活,或后者调节GPER的表达尚未被发现,以及这种相关性的功能结果。
    Prostate cancer (PCa) is the most prevalent cause of death in the male population worldwide. The G Protein-Coupled Estrogen Receptor (GPER) has been gaining relevance in the development of PCa. Hedgehog (Hh) pathway activation is associated with aggressiveness, metastasis, and relapse in PCa patients. To date, no studies have evaluated the crosstalk between the GPER and the Hh pathway along different group grades in PCa. We conducted an analysis of paraffin-embedded tissues derived from patients with different prognostic grade of PCa using immunohistochemistry. Expression and correlation between GPER and glioma associated oncogene homologue (GLI) transcriptional factors in the parenchyma and stroma of PCa tumors were evaluated. Our results indicate that GPER is highly expressed in the nucleus and increases with higher grade groups. Additionally, GPER\'s expression correlates with pGLI3 nuclear expression across different grade groups in PCa tissues; however, whether the receptor induces the activation of GLI transcriptional factors, or the latter modulate the expression of GPER is yet to be discovered, as well as the functional consequence of this correlation.
    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
    基因组结构的变化是生物体表型多样化的基础。氨基酸改变突变影响蛋白质的多效性功能,尽管对突变的蛋白质如何在现有的发育程序中适应知之甚少。在这里,我们研究了尼安德特人和丹尼索瓦人携带的GLI3转录因子(GLI3R1537C)变体的生物学效应,这是接近现代人类的灭绝的人类。R1537C不损害蛋白质稳定性或GLI3激活因子依赖性转录活性。相比之下,R1537C影响与发育过程相关的下游靶基因的调节。此外,携带尼安德特人/丹尼索瓦人GLI3突变的基因组编辑小鼠在骨骼形态上表现出各种改变。我们的数据表明,已灭绝的人源蛋白型GLI3有助于物种特异性解剖变异,在人类进化过程中,发展计划的放松约束是可以容忍的。
    Changes in genomic structures underlie phenotypic diversification in organisms. Amino acid-changing mutations affect pleiotropic functions of proteins, although little is known about how mutated proteins are adapted in existing developmental programs. Here we investigate the biological effects of a variant of the GLI3 transcription factor (GLI3R1537C) carried in Neanderthals and Denisovans, which are extinct hominins close to modern humans. R1537C does not compromise protein stability or GLI3 activator-dependent transcriptional activities. In contrast, R1537C affects the regulation of downstream target genes associated with developmental processes. Furthermore, genome-edited mice carrying the Neanderthal/Denisovan GLI3 mutation exhibited various alterations in skeletal morphology. Our data suggest that an extinct hominin-type GLI3 contributes to species-specific anatomical variations, which were tolerated by relaxed constraint in developmental programs during human evolution.
    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
    转录调节因子和hedgehog(Hh)信号通路效应子Gli3的异常表达已知会引发先天性疾病,最常影响中枢神经系统(CNS)和四肢。在胚胎发育过程中控制Gli3转录的基因组顺式调节景观的准确描绘对于解释与先天性缺陷相关的非编码变体至关重要。在这里,我们对分子进化速率缓慢的鱼类进行了比较基因组分析,以鉴定Gli3内含子间隔(CNE15-21)中七个先前未知的保守非编码元件(CNE)。斑马鱼的转基因实验表明,这些元素中的大多数驱动Gli3表达组织的活性,主要是鳍,CNS,还有心脏.这些CNE与人类疾病相关SNP的交叉将CNE15鉴定为推定的哺乳动物颅面增强剂,在脊椎动物中具有保守的活性,并可能受到与人类颅面形态相关的突变的影响。最后,在缓慢进化的鱼类(象鲨)中保守的附属物特异性CNE的比较功能解剖,但在硬骨鱼(CNE14/hs1586)中没有表明在羊膜和叶翅鱼分化之前,其他组织对肢体特异性的共同选择。这些结果揭示了内含子Gli3增强子的一个新子集,该子集出现在颌骨的共同祖先中,并且在进化多样化的过程中,其序列成分可能在其他物种中逐渐被修饰。本文受版权保护。保留所有权利。
    Abnormal expression of the transcriptional regulator and hedgehog (Hh) signaling pathway effector Gli3 is known to trigger congenital disease, most frequently affecting the central nervous system (CNS) and the limbs. Accurate delineation of the genomic cis-regulatory landscape controlling Gli3 transcription during embryonic development is critical for the interpretation of noncoding variants associated with congenital defects. Here, we employed a comparative genomic analysis on fish species with a slow rate of molecular evolution to identify seven previously unknown conserved noncoding elements (CNEs) in Gli3 intronic intervals (CNE15-21). Transgenic assays in zebrafish revealed that most of these elements drive activities in Gli3 expressing tissues, predominantly the fins, CNS, and the heart. Intersection of these CNEs with human disease associated SNPs identified CNE15 as a putative mammalian craniofacial enhancer, with conserved activity in vertebrates and potentially affected by mutation associated with human craniofacial morphology. Finally, comparative functional dissection of an appendage-specific CNE conserved in slowly evolving fish (elephant shark), but not in teleost (CNE14/hs1586) indicates co-option of limb specificity from other tissues prior to the divergence of amniotes and lobe-finned fish. These results uncover a novel subset of intronic Gli3 enhancers that arose in the common ancestor of gnathostomes and whose sequence components were likely gradually modified in other species during the process of evolutionary diversification.
    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
    对Hedgehog(HH)信号通路的转录反应主要由小鼠肢体中的GLI抑制调节。先前的研究表明BAF染色质重塑复合物在介导GLI抑制中的作用。与这种可能性一致,核心BAF复合物蛋白SMARCC1存在于大多数活跃的肢体增强剂,包括大多数GLI增强剂。然而,与降低染色质可及性的GLI抑制相反,SMARCC1在大多数增强剂中保持染色质可及性,包括那些受GLI约束的人。此外,SMARCC1在GLI调节的增强子上的结合独立于GLI3发生。与以前的研究一致,一些单独的GLI靶基因在Smarcc1条件性敲除中被错误调节,尽管大多数GLI靶基因不受影响。此外,SMARCC1对于介导HH突变肢芽中的组成型GLI抑制不是必需的。我们得出结论,SMARCC1不介导GLI3抑制,我们建议利用替代染色质重塑复合物。
    Transcriptional responses to the Hedgehog (HH) signaling pathway are primarily modulated by GLI repression in the mouse limb. Previous studies suggested a role for the BAF chromatin remodeling complex in mediating GLI repression. Consistent with this possibility, the core BAF complex protein SMARCC1 is present at most active limb enhancers including the majority of GLI enhancers. However, in contrast to GLI repression which reduces chromatin accessibility, SMARCC1 maintains chromatin accessibility at most enhancers, including those bound by GLI. Moreover, SMARCC1 binding at GLI-regulated enhancers occurs independently of GLI3. Consistent with previous studies, some individual GLI target genes are mis-regulated in Smarcc1 conditional knockouts, though most GLI target genes are unaffected. Moreover, SMARCC1 is not necessary for mediating constitutive GLI repression in HH mutant limb buds. We conclude that SMARCC1 does not mediate GLI3 repression, which we propose utilizes alternative chromatin remodeling complexes.
    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
    前列腺癌是一种依赖于雄激素刺激的疾病,因此通常用雄激素剥夺疗法(ADT)治疗。ADT最初非常成功;然而,患者不可避免地复发,此时癌症的生长与雄激素无关,被称为去势抵抗性前列腺癌(CRPC).CRPC通过各种机制发展,其中之一是雄激素受体(AR)信号通路与其他信号通路的串扰。一致地,先前的工作表明,雄激素剥夺诱导SHH信号,随后促进AR依赖性基因表达的激活以促进细胞生长。机械上,这种串扰涉及AR和SHH信号分量之间的物理相互作用,特别是GLI转录因子家族的蛋白质。因此,这些发现表明,SHH信号的激活可以在缺乏雄激素的情况下促进细胞生长的复发,最终导致向CRPC的进展。在这项研究中,我们已经在一个在Mediator亚基12(MED12)内具有遗传改变的前列腺癌子集中研究了这一机制.我们发现MED12的缺失促进GLI3靶基因的表达,其随后在不存在雄激素的情况下驱动细胞过度生长。因此,我们得出的结论是,MED12内的遗传改变通过过度激活的GLI3依赖的Sonichedgehog信号促进CRPC。
    Prostate cancer is a disease that depends on androgenic stimulation and is thus commonly treated with androgen deprivation therapy (ADT). ADT is highly successful initially; however, patients inevitably relapse at which point the cancer grows independently of androgens and is termed castration-resistant prostate cancer (CRPC). CRPC develops through various mechanisms, one of these being crosstalk of the androgen receptor (AR) signaling pathway with other signaling pathways. Congruently, prior work has shown that androgen deprivation induces SHH signaling, which subsequently promotes activation of AR-dependent gene expression to promote cell growth. Mechanistically, this crosstalk involves a physical interaction between AR and components of SHH signaling, specifically proteins of the GLI transcription factor family. These findings thus suggest that activation of SHH signaling could promote the recurrence of cell growth in the absence of androgens to ultimately lead to progression towards CRPC. In this study, we have investigated this mechanism in a subset of prostate cancer that harbors genetic alterations within the Mediator subunit 12 (MED12). We found that loss of MED12 promotes the expression of GLI3 target genes which subsequently drives excessive cell growth in the absence of androgens. Thus, we conclude that genetic alterations within MED12 promote CRPC through hyperactivated GLI3 dependent sonic hedgehog signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号