GNAS mutation

GNAS 突变
  • 文章类型: Journal Article
    腹膜假粘液瘤(PMP)是一种惰性恶性综合征。PMP的标准治疗是细胞减灭术加腹腔热化疗(CRS+HIPEC)。然而,高复发率和潜在的临床症状和体征是进一步改善临床结局的主要障碍。此外,由于广泛的腹膜内转移,晚期患者从CRS+HIPEC中获益甚微.PMP治疗的另一个挑战涉及PMP细胞分泌粘液的进行性硬化,通常由于编码鸟嘌呤核苷酸结合蛋白α亚基(GNAS)的基因中的激活突变而增加。因此,迫切需要开发其他PMP疗法。几种免疫相关疗法已经显示出希望,包括使用细菌来源的非特异性免疫原性试剂,放射性免疫治疗剂,和肿瘤细胞衍生的新抗原,但是尚未建立公认的免疫疗法。在这篇综述中,讨论了GNAS突变在促进粘蛋白分泌和疾病发展中的作用。此外,讨论了PMP微环境的免疫学特征和免疫相关治疗,以总结目前对疾病关键特征的理解,并促进免疫疗法的发展.
    Pseudomyxoma peritonei (PMP) is an indolent malignant syndrome. The standard treatment for PMP is cytoreductive surgery combined with intraperitoneal hyperthermic chemotherapy (CRS + HIPEC). However, the high recurrence rate and latent clinical symptoms and signs are major obstacles to further improving clinical outcomes. Moreover, patients in advanced stages receive little benefit from CRS + HIPEC due to widespread intraperitoneal metastases. Another challenge in PMP treatment involves the progressive sclerosis of PMP cell-secreted mucus, which is often increased due to activating mutations in the gene coding for guanine nucleotide-binding protein alpha subunit (GNAS). Consequently, the development of other PMP therapies is urgently needed. Several immune-related therapies have shown promise, including the use of bacterium-derived non-specific immunogenic agents, radio-immunotherapeutic agents, and tumor cell-derived neoantigens, but a well-recognized immunotherapy has not been established. In this review the roles of GNAS mutations in the promotion of mucin secretion and disease development are discussed. In addition, the immunologic features of the PMP microenvironment and immune-associated treatments are discussed to summarize the current understanding of key features of the disease and to facilitate the development of immunotherapies.
    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
    大约30%-40%的分泌生长激素的垂体腺瘤(GHPAs)在GNAS(刺激性G蛋白的α亚基)中具有体细胞激活突变。GNAS中的突变与较小且侵入性较小的肿瘤的临床特征相关。然而,GNAS突变在GHPAs侵袭中的作用尚不清楚.使用标准聚合酶链反应(PCR)测序程序在GHPA中检测GNAS突变。用RT-qPCR评估突变相关的母体表达基因3(MEG3)的表达。使用慢病毒表达系统在GH3细胞中操作MEG3。使用Transwell测定法测量细胞侵袭能力,和上皮间质转化(EMT)相关蛋白通过免疫荧光和蛋白质印迹进行定量。最后,使用肿瘤细胞异种移植小鼠模型来验证MEG3对肿瘤生长和侵袭性的影响。与具有野生型GNAS的小鼠相比,具有突变GNAS的小鼠的GHPAs的侵袭力明显降低。始终如一,突变表达GNAS的GH3细胞的侵袭力降低。MEG3在携带突变GNAS的GHPAs中以高水平独特表达。因此,MEG3上调抑制肿瘤细胞侵袭,反过来,MEG3下调增加肿瘤细胞侵袭。机械上,GNAS突变抑制GHPAs中的EMT。突变的GNAS细胞中的MEG3通过Wnt/β-catenin信号通路的失活阻止细胞侵袭,进一步在体内验证。我们的数据表明,GNAS突变可能通过MEG3/Wnt/β-catenin信号通路的激活来调节EMT,从而抑制GHPAs中的细胞侵袭。
    Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/β-catenin signaling pathway.
    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
    细胞粘液瘤是一种良性软组织肿瘤,通常与GNAS突变相关,在形态上可能类似于低度粘液纤维肉瘤。这项研究旨在鉴定细胞粘液瘤的未描述的甲基化谱,并将其与粘液纤维肉瘤进行比较。我们对20个细胞粘液瘤和9个粘液纤维肉瘤进行了分子分析,并使用基于甲基化的DKFZ肉瘤分类器分析了结果。总共90%的细胞粘液瘤具有GNAS突变(以前没有描述过四个基因座)。在所有粘液纤维肉瘤中都发现了拷贝数变异,但在没有细胞粘液瘤中发现了拷贝数变异。在分类器中,细胞粘液瘤均未达到0.9阈值。无监督t-SNE分析表明,细胞粘液瘤形成自己的簇,与粘纤维肉瘤不同。我们的研究表明,在形态学和免疫组织化学不足以区分细胞粘液瘤和粘液纤维肉瘤的情况下,分子分析的诊断潜力和局限性。特别是关于GNAS野生型肿瘤。DKFZ肉瘤分类器仅提供了一个粘液纤维肉瘤病例的有效预测;可以通过用更多的病例训练工具来改善这种限制。此外,分类器应该引入更广泛的间充质肿瘤,包括细胞粘液瘤等良性肿瘤,我们证明了其独特的甲基化模式。
    Cellular myxoma is a benign soft tissue tumor frequently associated with GNAS mutation that may morphologically resemble low-grade myxofibrosarcoma. This study aimed to identify the undescribed methylation profile of cellular myxoma and compare it to myxofibrosarcoma. We performed molecular analysis on twenty cellular myxomas and nine myxofibrosarcomas and analyzed the results using the methylation-based DKFZ sarcoma classifier. A total of 90% of the cellular myxomas had GNAS mutations (four loci had not been previously described). Copy number variations were found in all myxofibrosarcomas but in none of the cellular myxomas. In the classifier, none of the cellular myxomas reached the 0.9 threshold. Unsupervised t-SNE analysis demonstrated that cellular myxomas form their own clusters, distinct from myxofibrosarcomas. Our study shows the diagnostic potential and the limitations of molecular analysis in cases where morphology and immunohistochemistry are not sufficient to distinguish cellular myxoma from myxofibrosarcoma, particularly regarding GNAS wild-type tumors. The DKFZ sarcoma classifier only provided a valid prediction for one myxofibrosarcoma case; this limitation could be improved by training the tool with a more considerable number of cases. Additionally, the classifier should be introduced to a broader spectrum of mesenchymal neoplasms, including benign tumors like cellular myxoma, whose distinct methylation pattern we demonstrated.
    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
    纤维发育不良(FD)是一种罕见的骨骼疾病,其特征是用良性纤维骨组织代替正常骨骼。缺乏合适的研究模型阻碍了我们对病理生理学和治疗选择的理解的发展。在这项研究中,我们开发了一种体外器官型模型,能够概括FD的关键内在和表型特性。最初,从患者病变组织中分离的单个细胞的转录组学分析揭示了病变内分子和细胞异质性。利用这些见解,我们使用从患者FD病变获得的原代细胞建立了患者来源的类器官(PDO).PDO的评估证明了在FD病变中观察到的纤维化相关组成细胞类型和转录特征的保留。此外,PDO保留了FD特有的基因组和代谢改变的不同星座。组织学评估进一步证实了PDO的保真度,以概括FD的重要表型特征,强调了其病理生理相关性。我们的发现代表了该领域的有意义的进展,因为它们为三维背景下罕见骨病变的体外建模开辟了可能性,并且可能标志着为研究和治疗研究创建个性化平台的第一步。
    Fibrous dysplasia (FD) is a rare bone disorder characterized by the replacement of normal bone with benign fibro-osseous tissue. Developments in our understanding of the pathophysiology and treatment options are impeded by the lack of suitable research models. In this study, we developed an in vitro organotypic model capable of recapitulating key intrinsic and phenotypic properties of FD. Initially, transcriptomic profiling of individual cells isolated from patient lesional tissues unveiled intralesional molecular and cellular heterogeneity. Leveraging these insights, we established patient-derived organoids (PDOs) using primary cells obtained from patient FD lesions. Evaluation of PDOs demonstrated preservation of fibrosis-associated constituent cell types and transcriptional signatures observed in FD lesions. Additionally, PDOs retained distinct constellations of genomic and metabolic alterations characteristic of FD. Histological evaluation further corroborated the fidelity of PDOs in recapitulating important phenotypic features of FD that underscore their pathophysiological relevance. Our findings represent meaningful progress in the field, as they open up the possibility for in vitro modeling of rare bone lesions in a three-dimensional context and may signify the first step towards creating a personalized platform for research and therapeutic studies.
    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:腹膜假粘液瘤(PMP)是一种疾病,其特征是由腹腔肿瘤产生的腹膜内粘液性腹水的进行性积累。由于PMP患者的预后仍然不令人满意,开发有效的治疗药物是一个紧迫的问题。PMP的遗传分析已经阐明了GNAS和/或KRAS的频繁激活。然而,PMPs涉及全球表观遗传改变的报道尚未出现.
    方法:为了阐明15个PMP肿瘤的遗传背景,我们使用AmpliSeq癌症热点面板v2进行了遗传分析。我们使用包含总共865,918个探针的甲基化EPIC阵列BeadChip(Infinium850k)进一步研究了15种肿瘤和8种非癌性结肠上皮细胞中的全局DNA甲基化。
    结果:这是世界上有关PMPs的全面DNA甲基化谱的第一份报告。我们澄清了15个PMPs可以分为至少两种表观基因型,独特的甲基化表观基因型(UME)和正常样甲基化表观基因型(NLME),与神经元发育和突触信号相关的基因可能参与了PMPs的发育。此外,我们在15个PMPs中鉴定了一组高甲基化标记基因如HOXD1和TSPYL5。
    结论:这些发现可能有助于了解PMP的分子机制,并有助于开发这种危及生命的疾病的治疗策略。
    BACKGROUND: Pseudomyxoma peritonei (PMP) is a disease characterized by progressive accumulation of intraperitoneal mucinous ascites produced by neoplasms in the abdominal cavity. Since the prognosis of patients with PMP remains unsatisfactory, the development of effective therapeutic drug(s) is a matter of pressing concern. Genetic analyses of PMP have clarified the frequent activation of GNAS and/or KRAS. However, the involvement of global epigenetic alterations in PMPs has not been reported.
    METHODS: To clarify the genetic background of the 15 PMP tumors, we performed genetic analysis using AmpliSeq Cancer HotSpot Panel v2. We further investigated global DNA methylation in the 15 tumors and eight noncancerous colonic epithelial tissues using MethylationEPIC array BeadChip (Infinium 850k) containing a total of 865,918 probes.
    RESULTS: This is the first report of comprehensive DNA methylation profiles of PMPs in the world. We clarified that the 15 PMPs could be classified into at least two epigenotypes, unique methylation epigenotype (UME) and normal-like methylation epigenotype (NLME), and that genes associated with neuronal development and synaptic signaling may be involved in the development of PMPs. In addition, we identified a set of hypermethylation marker genes such as HOXD1 and TSPYL5 in the 15 PMPs.
    CONCLUSIONS: These findings may help the understanding of the molecular mechanism(s) of PMP and contribute to the development of therapeutic strategies for this life-threatening disease.
    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
    纤维发育不良(FD)是一种罕见的,非遗传性骨骼疾病,其特征是慢性非肿瘤性纤维组织积聚代替健康骨骼。无数因素与其发病和进展有关。细胞-细胞信号网络和响应输出的扰动导致构建块中断,不连贯的多层次组织,矿化组织中刚性结构基序的丢失是已确定参与FD诱导的因素。近年来,对FD独特生物学的新见解正在改变我们对其病理学的理解,疾病的自然话语,和治疗前景。在这里,我们基于现有的知识和最近的发现来回顾临床,病因学,和FD的组织学特征,并讨论了FD表现的已知和潜在机制。随后,我们以乐观的态度结束,我们强调了旨在阻止或改善疾病进展的新兴治疗方法.
    Fibrous dysplasia (FD) is a rare, non-hereditary skeletal disorder characterized by its chronic course of non-neoplastic fibrous tissue buildup in place of healthy bone. A myriad of factors have been associated with its onset and progression. Perturbation of cell-cell signaling networks and response outputs leading to disrupted building blocks, incoherent multi-level organization, and loss of rigid structural motifs in mineralized tissues are factors that have been identified to participate in FD induction. In more recent years, novel insights into the unique biology of FD are transforming our understandings of its pathology, natural discourse of the disease, and treatment prospects. Herein, we built upon existing knowledge with recent findings to review clinical, etiologic, and histological features of FD and discussed known and potential mechanisms underlying FD manifestations. Subsequently, we ended on a note of optimism by highlighting emerging therapeutic approaches aimed at either halting or ameliorating disease progression.
    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
    髓母细胞瘤(MB)是最常见的恶性脑肿瘤,发生在儿童时期,在成人中很少见。根据转录组概况,MB目前分为四个主要分子组,反映了相当大的生物异质性:WNT激活,SHH激活,第3组和第4组。最近,DNA甲基化分析允许鉴定与不同临床病理和分子特征相关的四个主要分子组中的其他亚组。异柠檬酸脱氢酶-1和2(IDH1和IDH2)突变已在几种肿瘤中得到描述。包括神经胶质瘤,而在MB中很少报告,也没有常规调查。通过磁共振波谱(MRS),我们明确评估了体内代谢物D-2-羟基戊二酸(2HG)的存在,IDH1和IDH2突变的标记,在成人MB的情况下。免疫表型检查和甲基化分析分配了MB的诊断,SHH-A子类,和分子测试表明存在非规范体细胞IDH1(p。R132C)突变和额外的GNAS突变,也很少用MB描述。据我们所知,这是报道的首例同时携带两种突变的MB病例.值得注意的是,肿瘤表现出异质性表型,肿瘤成分表现出神经胶质分化,具有强大的GFAP表达式,以及具有常规MB特征和选择性存在GNAS突变的组件,表明两个不同的主要肿瘤亚克隆共存。这些发现提请注意需要对MB进行更深入的遗传表征,为了深入了解他们的生物学,改善患者的分层和临床管理。此外,我们的结果强调了在非神经胶质肿瘤中进行MRS对于识别IDH突变的重要性.使用通量分子谱分析和先进的医学成像肯定会增加具有罕见分子改变的肿瘤实体被识别的频率。这些发现是否具有任何特定的治疗意义或预后相关性需要进一步研究。
    Medulloblastoma (MB) is the most common malignant brain tumor occurring in childhood and rarely found in adults. Based on transcriptome profile, MB are currently classified into four major molecular groups reflecting a considerable biological heterogeneity: WNT-activated, SHH-activated, group 3 and group 4. Recently, DNA methylation profiling allowed the identification of additional subgroups within the four major molecular groups associated with different clinic-pathological and molecular features. Isocitrate dehydrogenase-1 and 2 (IDH1 and IDH2) mutations have been described in several tumors, including gliomas, while in MB are rarely reported and not routinely investigated. By means of magnetic resonance spectroscopy (MRS), we unequivocally assessed the presence the oncometabolite D-2-hydroxyglutarate (2HG), a marker of IDH1 and IDH2 mutations, in a case of adult MB. Immunophenotypical work-up and methylation profiling assigned the diagnosis of MB, subclass SHH-A, and molecular testing revealed the presence of the non-canonical somatic IDH1(p.R132C) mutation and an additional GNAS mutation, also rarely described in MB. To the best of our knowledge, this is the first reported case of MB simultaneously harboring both mutations. Of note, tumor exhibited a heterogeneous phenotype with a tumor component displaying glial differentiation, with robust GFAP expression, and a component with conventional MB features and selective presence of GNAS mutation, suggesting co-existence of two different major tumor subclones. These findings drew attention to the need for a deeper genetic characterization of MB, in order to get insights into their biology and improve stratification and clinical management of the patients. Moreover, our results underlined the importance of performing MRS for the identification of IDH mutations in non-glial tumors. The use of throughput molecular profiling analysis and advanced medical imaging will certainly increase the frequency with which tumor entities with rare molecular alterations will be identified. Whether these findings have any specific therapeutic implications or prognostic relevance requires further investigations.
    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
    儿童原发性肾上腺功能不全具有非特异性和广泛的临床特征,因此其病因的诊断既复杂又具有挑战性。虽然先天性肾上腺增生是最常见的原因,已经确定了越来越多的其他遗传原因。GNAS突变是原发性肾上腺皮质功能不全的罕见原因,容易被忽视。本文首次报道一例新生儿GNAS突变导致原发性肾上腺功能不全的病例。
    一名男孩在产后10天被诊断为先天性甲状腺功能减退症,并立即接受治疗。他还患有持续性高钾血症和低钠血症,伴有促肾上腺皮质激素升高。出生后70天,他因涉嫌先天性肾上腺增生被转到我们医院。体格检查发现除生长迟缓外无其他异常。实验室检查显示醛固酮增加和皮质醇正常,17-羟基孕酮,和雄烯二酮水平。甲状旁腺激素异常升高伴有正常的血钙。基因评估发现了一个从头,杂合c.432+1G>GNAS中的一个变异体。
    我们报道这个病例是为了强调GNAS突变是原发性肾上腺功能不全的一个不寻常的原因。原发性甲状腺功能减退症和/或假性甲状旁腺功能减退症的组合将为这种情况提供诊断线索。
    BACKGROUND: Primary adrenal insufficiency in children has non-specific and extensive clinical features, so the diagnosis of its etiology is complex and challenging. Although congenital adrenal hyperplasia is the most common cause, more and more other genetic causes have been identified. GNAS mutation is easily overlooked as a rare cause of primary adrenal insufficiency. Here we firstly report a neonatal case of primary adrenal insufficiency caused by GNAS mutation.
    METHODS: A boy was diagnosed with congenital hypothyroidism 10 days post-partum and treated immediately. He also had persistent hyperkalaemia and hyponatraemia with elevated adrenocorticotropic hormone. At 70 days after birth, he was transferred to our hospital on suspicion of congenital adrenal hyperplasia. Physical examination found no other abnormalities except for growth retardation. Laboratory examination revealed increased aldosterone and normal cortisol, 17-hydroxyprogesterone, and androstenedione levels. Abnormally elevated parathyroid hormone was accompanied by normal blood calcium. Genetic assessment found a de novo, heterozygous c.432 + 1G > A variant in GNAS.
    CONCLUSIONS: We report this case to highlight that GNAS mutation is an unusual cause of primary adrenal insufficiency. The combination of primary hypothyroidism and /or pseudohypoparathyroidism will provide diagnostic clues to this condition.
    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
    UNASSIGNED: Although surgical resection can cure the majority of meningiomas, there are still approximately 20% of patients suffering from an aggressive course with recurrence or progression. In this study, we reported a novel GNAS mutation and 1p/22q co-deletion responding to sunitinib in a patient with multiple recurrent meningiomas.
    UNASSIGNED: A 53-year-old woman with meningioma was hospitalized due to postoperative tumor progression for 3 weeks. WHO grade I meningioma was pathologically diagnosed after the first three surgeries, but the second recurrence occurred approximately 3 years following the third surgery. Next-generation sequencing was performed on the first two recurrent samples. GNAS mutations and 1p/22q co-deletion were both identified, and amplification at 17q and chromosome 19 was also found in the second recurrent sample, based on which WHO grade II/III meningioma was diagnosed. The lesion in the left cerebellopontine angle area enlarged after use of radiotherapy combined with temozolomide chemotherapy for 2 months. When sunitinib was added, the residual lesions began to lessen and continuously reduced.
    UNASSIGNED: This typical case suggested that timely molecular diagnosis for refractory meningiomas contributed to guiding the molecular classification and clinicians to make more reasonable individualized therapeutic regimens, consequently benefiting the patients. This case report also highlighted the potential role of sunitinib in the treatment of refractory meningiomas.
    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
    BACKGROUND: Ectopic pancreas is basically a benign disease and is not always necessary to be removed. However, all types of neoplasms occurring in the normal pancreas such as ductal adenocarcinomas and intraductal papillary mucinous neoplasms (IPMNs) may develop even within ectopic pancreas. We recently encountered an extremely rare case of ectopic pancreas in the gastric antrum associated with IPMN possessing a GNAS mutation.
    METHODS: A 71-year-old Japanese woman complained of epigastric pain. Computed tomography and upper gastrointestinal endoscopy showed an intramural cystic mass in the antrum of the stomach. Endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) biopsy did not give a definitive diagnosis, and the patient underwent resection of the lesion. Histology of the resected specimen showed that the gastric intramural lesion was ectopic pancreas. Moreover, the lesion contained dilated duct components with tubulo-villous epithelial proliferation consistent with pancreatic IPMN. Since the covering epithelial cells had highly atypical nuclei, the lesion was diagnosed as IPMN with high grade dysplasia. Immunohistochemistry showed that the IPMN component showed to be MUC2-, MUC5AC-, and CDX2-positive but MUC1- and MUC6-negative. Mutational analyses using genomic DNA revealed that the IPMN component had a mutation of GNAS at exon 8 (Arg201Cys).
    CONCLUSIONS: We finally diagnosed this case as gastric ectopic pancreas accompanied by intestinal type IPMN with high grade dysplasia possessing GNAS mutation. Although there were 17 cases of ectopic pancreas with IPMN including 6 cases of gastric ones reported in the English literature, this is the first case of ectopic pancreas with IPMN which was proved to have GNAS mutation. Intimate preoperative examinations including imaging analyses and EUS-FNA biopsy/cytology are recommended to decide whether the lesion has to be resected or not even if they are not effective for getting the right diagnosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号