KDM5D

KDM5D
  • 文章类型: Clinical Study
    背景:克罗恩病(CD)患者对ustekinumab(UST)治疗的反应存在差异,但根本原因仍然未知。我们的目的是研究免疫细胞的参与,并确定可以预测CD患者对白介素(IL)12/23抑制剂的反应的潜在生物标志物。
    方法:GSE207022数据集,其中包括CD队列中的54名非响应者和9名UST响应者,被分析。鉴定差异表达基因(DEGs)并进行基因本体论(GO)和京都基因和基因组百科全书(KEGG)途径分析。使用最小绝对收缩和选择算子(LASSO)回归来筛选最强大的集线器基因。进行受试者工作特征(ROC)曲线分析以评估这些基因的预测性能。使用单样品基因组富集分析(ssGSEA)来估计免疫细胞类型的比例。对这些显著改变的基因进行聚类分析,形成免疫细胞相关的浸润。为了验证候选人的可靠性,在前瞻性队列中使用UST作为一线生物制剂的患者被纳入作为独立的验证数据集.
    结果:在综合数据集中确定了总共99个DEG。GO和KEGG分析显示CD患者的免疫应答途径显著富集。13个基因(SOCS3,CD55,KDM5D,IGFBP5,LCN2,SLC15A1,XPNPEP2,HLA-DQA2,HMGCS2,DDX3Y,ITGB2,CDKN2B和HLA-DQA1),主要与有反应的患者和无反应的患者有关,被识别并包括在LASSO分析中。这些基因准确地预测了治疗反应,曲线下面积(AUC)为0.938。在无反应个体中,1型T辅助细胞(Th1)细胞极化相对较强。在Th1细胞与LCN2和KDM5D基因之间观察到正连接。此外,我们采用独立的验证数据集和早期实验验证来验证LCN2和KDM5D基因作为有效的预测标记.
    结论:Th1细胞极化是CD患者对UST治疗无反应的重要原因。LCN2和KDM5D可用作预测标志物以有效识别无应答患者。
    背景:试用注册号:NCT05542459;注册日期:2022-09-14;URL:https://www。
    结果:政府。
    BACKGROUND: Variations exist in the response of patients with Crohn\'s disease (CD) to ustekinumab (UST) treatment, but the underlying cause remains unknown. Our objective was to investigate the involvement of immune cells and identify potential biomarkers that could predict the response to interleukin (IL) 12/23 inhibitors in patients with CD.
    METHODS: The GSE207022 dataset, which consisted of 54 non-responders and 9 responders to UST in a CD cohort, was analyzed. Differentially expressed genes (DEGs) were identified and subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Least absolute shrinkage and selection operator (LASSO) regression was used to screen the most powerful hub genes. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performances of these genes. Single-sample Gene Set Enrichment Analysis (ssGSEA) was used to estimate the proportions of immune cell types. These significantly altered genes were subjected to cluster analysis into immune cell-related infiltration. To validate the reliability of the candidates, patients prescribed UST as a first-line biologic in a prospective cohort were included as an independent validation dataset.
    RESULTS: A total of 99 DEGs were identified in the integrated dataset. GO and KEGG analyses revealed significant enrichment of immune response pathways in patients with CD. Thirteen genes (SOCS3, CD55, KDM5D, IGFBP5, LCN2, SLC15A1, XPNPEP2, HLA-DQA2, HMGCS2, DDX3Y, ITGB2, CDKN2B and HLA-DQA1), which were primarily associated with the response versus nonresponse patients, were identified and included in the LASSO analysis. These genes accurately predicted treatment response, with an area under the curve (AUC) of 0.938. T helper cell type 1 (Th1) cell polarization was comparatively strong in nonresponse individuals. Positive connections were observed between Th1 cells and the LCN2 and KDM5D genes. Furthermore, we employed an independent validation dataset and early experimental verification to validate the LCN2 and KDM5D genes as effective predictive markers.
    CONCLUSIONS: Th1 cell polarization is an important cause of nonresponse to UST therapy in patients with CD. LCN2 and KDM5D can be used as predictive markers to effectively identify nonresponse patients.
    BACKGROUND: Trial registration number: NCT05542459; Date of registration: 2022-09-14; URL: https://www.
    RESULTS: gov .
    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
    发育中的人脑中的性别差异主要归因于荷尔蒙的影响。然而,最近,遗传差异及其对发育中的神经系统的影响引起了越来越多的关注。为了了解神经发育中遗传驱动的性二态,我们在男性和女性人类胚胎干细胞系(hESC)的体外分化模型中研究了全基因组基因表达,独立于人类性激素的影响。四个雄性和四个雌性来源的hESC品系在37天内分化成混合神经元群体。对大量RNA测序数据进行差异基因表达和基因集富集分析。虽然在所有细胞系中相似的分化倾向证明了我们的分化方案的稳健性和可重复性,在第0天,我们发现未分化的ESCs中已经存在性别偏倚的基因表达,但在分化37天后最为明显.男性和女性细胞系表现出参与神经发育的基因的性别偏倚表达,这表明性别会影响分化轨迹。有趣的是,发现性别差异的最大贡献来自男性转录组,涉及Y染色体和常染色体基因。我们提出了13个性别偏向的候选基因(10个在雄性细胞系中上调,3个在雌性细胞系中上调),这些基因可能会影响神经元的发育。此外,我们证实了通过Y同源物逃避X染色体失活的X/Y同源物的基因剂量补偿,并确定了在神经元发育过程中男性hESC中Y连锁脱甲基酶UTY和KDM5D的显著过表达,证实了以前在神经干细胞中的结果。我们的结果表明,遗传性别差异影响神经元分化轨迹,这最终可能导致人类大脑发育过程中的性别偏见。
    Sex differences in the developing human brain are primarily attributed to hormonal influence. Recently however, genetic differences and their impact on the developing nervous system have attracted increased attention. To understand genetically driven sexual dimorphisms in neurodevelopment, we investigated genome-wide gene expression in an in vitro differentiation model of male and female human embryonic stem cell lines (hESC), independent of the effects of human sex hormones. Four male and four female-derived hESC lines were differentiated into a population of mixed neurons over 37 days. Differential gene expression and gene set enrichment analyses were conducted on bulk RNA sequencing data. While similar differentiation tendencies in all cell lines demonstrated the robustness and reproducibility of our differentiation protocol, we found sex-biased gene expression already in undifferentiated ESCs at day 0, but most profoundly after 37 days of differentiation. Male and female cell lines exhibited sex-biased expression of genes involved in neurodevelopment, suggesting that sex influences the differentiation trajectory. Interestingly, the highest contribution to sex differences was found to arise from the male transcriptome, involving both Y chromosome and autosomal genes. We propose 13 sex-biased candidate genes (10 upregulated in male cell lines and 3 in female lines) that are likely to affect neuronal development. Additionally, we confirmed gene dosage compensation of X/Y homologs escaping X chromosome inactivation through their Y homologs and identified a significant overexpression of the Y-linked demethylase UTY and KDM5D in male hESC during neuron development, confirming previous results in neural stem cells. Our results suggest that genetic sex differences affect neuronal differentiation trajectories, which could ultimately contribute to sex biases during human brain development.
    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:m6A修饰目前被认为是维持癌细胞稳态的RNA功能的主要驱动因素。长链非编码(Lnc)RNA控制细胞增殖,在结直肠癌(CRC)的发生和发展中起重要作用。ZCCHC4是一种新发现的m6A甲基转移酶,其在肿瘤中的作用和机制尚未阐明。
    方法:使用EpiQuikm6ARNA甲基化试剂盒检测六种消化道肿瘤中总RNAm6A的水平。采用Kaplan-Meier法和受试者工作特征曲线评价新发现的m6A甲基转移酶的预后和诊断价值,ZCCHC4,在CRC中。使用功能增益和功能丧失实验研究了体外和体内对CRC生长的影响。使用RIP研究了CRC中ZCCHC4上调的表观遗传机制,MeRIP-seq,RNA下拉,和动物实验。
    结果:我们报道了ZCCHC4-LncRNAGHRLOS-KDM5D轴在体外和体内调节CRC的生长。我们发现ZCCHC4在原发性CRC样本中上调,可以预测CRC患者的不良临床结局。机械上,ZCCHC4下调LncRNAGHRLOS以促进CRC肿瘤发生。作为LncRNAGHRLOS的下游分子,KDM5D直接控制CRC细胞增殖,迁移,和入侵。
    结论:本研究提示ZCCHC4轴有助于CRC的肿瘤发生和进展,ZCCHC4可能是该恶性肿瘤的潜在生物标志物。
    BACKGROUND: m6A modification is currently recognized as a major driver of RNA function that maintains cancer cell homeostasis. Long non-coding (Lnc) RNAs control cell proliferation and play an important role in the occurrence and progression of colorectal cancer (CRC). ZCCHC4 is a newly discovered m6A methyltransferase whose role and mechanism in tumors have not yet been elucidated.
    METHODS: The EpiQuik m6A RNA methylation kit was used to detect the level of total RNA m6A in six types of digestive tract tumors. The Kaplan-Meier method and receiver operating characteristic curve were used to evaluate the prognostic and diagnostic value of the newly discovered m6A methyltransferase, ZCCHC4, in CRC. The effects on CRC growth in vitro and in vivo were studied using gain- and loss-of-function experiments. The epigenetic mechanisms underlying ZCCHC4 upregulation in CRC were studied using RIP, MeRIP-seq, RNA pull-down, and animal experiments.
    RESULTS: We reported that the ZCCHC4-LncRNAGHRLOS-KDM5D axis regulates the growth of CRC in vitro and in vivo. We found that ZCCHC4 was upregulated in primary CRC samples and could predict adverse clinical outcomes in patients with CRC. Mechanistically, ZCCHC4 downregulated LncRNAGHRLOS to promote CRC tumorigenesis. As a downstream molecule of LncRNAGHRLOS, KDM5D directly controls CRC cell proliferation, migration, and invasion.
    CONCLUSIONS: This study suggests that the ZCCHC4 axis contributes to the tumorigenesis and progression of CRC and that ZCCHC4 may be a potential biomarker for this malignancy.
    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
    由于缺乏可靶向的驱动改变,有限数量的肺鳞状细胞癌(SCC)患者在临床上受益于分子靶向药物。我们旨在了解SCC中赖氨酸特异性脱甲基酶5D(KDM5D)拷贝数丢失的患病率和临床意义,并探讨其作为共济失调毛细血管扩张症和Rad3相关(ATR)抑制剂治疗的预测性生物标志物的潜力。我们使用荧光原位杂交评估了173例男性患者手术切除的SCC中的KDM5D拷贝数损失。在173名患者中的75名(43%)中检测到KDM5D拷贝数丢失。转录起始位点(TSS)的全基因组表达谱来自17个SCC,进行了基因表达测定的cap分析,揭示了在具有KDM5D拷贝数丢失的肿瘤中上调的基因与“细胞周期”有关,而在KDM5D拷贝数丢失的肿瘤中下调的基因与“免疫反应”相关。临床病理,具有KDM5D拷贝数丢失的SCCs与晚期病理阶段(p=0.0085)和高基质含量(p=0.0254)相关。多重荧光免疫组织化学显示,具有KDM5D拷贝数丢失的SCC中肿瘤浸润性CD8/T-betT细胞的数量低于野生型肿瘤。总之,约40%的男性SCC患者出现KDM5D拷贝数丢失.显示这种独特表型的患者的肿瘤可以是“冷肿瘤”,其特征是缺乏肿瘤T细胞浸润,通常对免疫疗法没有反应。因此,他们可能是ATR抑制剂试验的候选人.
    A limited number of patients with lung squamous cell carcinoma (SCC) benefit clinically from molecular targeted drugs because of a lack of targetable driver alterations. We aimed to understand the prevalence and clinical significance of lysine-specific demethylase 5D (KDM5D) copy number loss in SCC and explore its potential as a predictive biomarker for ataxia-telangiectasia and Rad3-related (ATR) inhibitor treatment. We evaluated KDM5D copy number loss in 173 surgically resected SCCs from male patients using fluorescence in situ hybridization. KDM5D copy number loss was detected in 75 of the 173 patients (43%). Genome-wide expression profiles of the transcription start sites (TSSs) were obtained from 17 SCCs, for which the cap analysis of gene expression assay was performed, revealing that upregulated genes in tumors with the KDM5D copy number loss are associated with \'cell cycle\', whereas downregulated genes in tumors with KDM5D copy number loss were associated with \'immune response\'. Clinicopathologically, SCCs with KDM5D copy number loss were associated with late pathological stage (p = 0.0085) and high stromal content (p = 0.0254). Multiplexed fluorescent immunohistochemistry showed that the number of tumor-infiltrating CD8+ /T-bet+ T cells was lower in SCCs with KDM5D copy number loss than in wild-type tumors. In conclusion, approximately 40% of the male patients with SCC exhibited KDM5D copy number loss. Tumors in patients who show this distinct phenotype can be \'cold tumors\', which are characterized by the paucity of tumor T-cell infiltration and usually do not respond to immunotherapy. Thus, they may be candidates for trials with ATR inhibitors.
    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
    Head and neck squamous cell carcinoma (HNSCC) is a major contributor to cancer incidence globally and is currently managed by surgical resection followed by adjuvant chemoradiotherapy. However, local recurrence is the major cause of mortality, indicating the emergence of drug-tolerant persister cells. A specific histone demethylase, namely lysine-specific demethylase 5D (KDM5D), is overexpressed in diverse types of cancers and involved in cancer cell cycle regulation. However, the role of KDM5D in the development of cisplatin-tolerant persister cells remains unexplored. Here, we demonstrated that KDM5D contributes to the development of persister cells. Aurora Kinase B (AURKB) disruption affected the vulnerability of persister cells in a mitotic catastrophe-dependent manner. Comprehensive in silico, in vitro, and in vivo experiments were performed. KDM5D expression was upregulated in HNSCC tumor cells, cancer stem cells, and cisplatin-resistant cells with biologically distinct signaling alterations. In an HNSCC cohort, high KDM5D expression was associated with a poor response to platinum treatment and early disease recurrence. KDM5D knockdown reduced the tolerance of persister cells to platinum agents and caused marked cell cycle deregulation, including the loss of DNA damage prevention, and abnormal mitosis-enhanced cell cycle arrest. By modulating mRNA levels of AURKB, KDM5D promoted the generation of platinum-tolerant persister cells in vitro, leading to the identification of the KDM5D/AURKB axis, which regulates cancer stemness and drug tolerance of HNSCC. Treatment with an AURKB inhibitor, namely barasertib, resulted in a lethal consequence of mitotic catastrophe in HNSCC persister cells. The cotreatment of cisplatin and barasertib suppressed tumor growth in the tumor mouse model. Thus, KDM5D might be involved in the development of persister cells, and AURKB disruption can overcome tolerance to platinum treatment in HNSCC.
    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
    现在,资助机构认识到并授权在生物医学研究中考虑性别的必要性。在实验室啮齿动物中,比如老鼠,性别鉴定通常在解剖学上或通过使用多重或单纯形PCR技术对基因组DNA进行基因分型。在这里,我们提出了一种简单的基于RT-PCR的方法,靶向Kdm5c和Kdm5d来确定小鼠cDNA样品中的遗传性别,允许回顾性性别确定。
    The need to take sex into account in biomedical research is now recognized and mandated by funding institutions. In laboratory rodents, such as mice, sexing is usually performed anatomically or by genotyping using multiplex or simplex PCR techniques on genomic DNA. Here we present a simple RT-PCR-based method targeting Kdm5c and Kdm5d to determine genetic sex in mouse cDNA samples, allowing for retrospective sex determination.
    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
    女性在衰老过程中与骨量减少和发生骨不连骨折的风险增加有关。荷尔蒙因素和细胞内在机制被认为驱动这些性二态,虽然潜在的分子机制仍然是一个争论的问题。这里,我们观察到雌性大鼠的颅骨恢复能力下降,而人类成年神经嵴来源的干细胞(NCSC)的性二形成骨分化明显.除了促成骨调节因子的表达升高之外,全球转录组学显示,赖氨酸脱甲基酶5D(KDM5D)在分化雄性NCSC中高度上调。siRNA导致的功能丧失或KDM5D的药理学抑制显著降低了雄性NCSC的成骨分化能力。总之,我们证明颅面成骨分化是性别二态的,KDM5D的表达是加速男性成骨分化的先决条件,强调性别特异性差异的分析作为治疗骨缺损的关键参数。
    Female sex is increasingly associated with a loss of bone mass during aging and an increased risk of developing nonunion fractures. Hormonal factors and cell-intrinsic mechanisms are suggested to drive these sexual dimorphisms, although underlying molecular mechanisms are still a matter of debate. Here, we observed a decreased capacity of calvarial bone recovery in female rats and a profound sexually dimorphic osteogenic differentiation in human adult neural crest-derived stem cells (NCSCs). Next to an elevated expression of pro-osteogenic regulators, global transcriptomics revealed Lysine Demethylase 5D (KDM5D) to be highly upregulated in differentiating male NCSCs. Loss of function by siRNA or pharmacological inhibition of KDM5D significantly reduced the osteogenic differentiation capacity of male NCSCs. In summary, we demonstrated craniofacial osteogenic differentiation to be sexually dimorphic with the expression of KDM5D as a prerequisite for accelerated male osteogenic differentiation, emphasizing the analysis of sex-specific differences as a crucial parameter for treating bone defects.
    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: The administration of docetaxel chemotherapy is one therapeutic option to delay disease progression and increase overall survival in metastatic castration resistant prostate cancer (mCRPC). However, about 15% of patients are primary resistant to chemotherapy and hence would benefit from an alternative mCRPC treatment. Despite intensive research, there are no robust clinical validated biomarkers to predict mCRPC therapy response. Thus, the aim of the study was to determine KDM5D expression in archival radical prostatectomy specimens of patients medicated with docetaxel at time of mCRPC development in order to correlate KMD5D expression with treatment response.
    UNASSIGNED: We used in situ hybridization (ISH) (RNA scope 2.5 HD) to determine KDM5D expression in tissue samples of 28 prostate cancer patients. KDM5D status was correlated to chemotherapy response (PSA and radiographic response).
    UNASSIGNED: Data revealed that KDM5D is significantly overexpressed in tumor cells (P<0.0001) but also in benign cells (P<0.02) of those patients who responded to chemotherapy compared to non-responders.
    UNASSIGNED: To summarize, KDM5D is a promising novel biomarker predicting response to docetaxel chemotherapy already at the time of localized disease and thus potentially avoiding metastatic biopsies in the mCRPC stage of 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
    Colorectal cancer (CRC) remains the most frequently diagnosed malignancy and also a major contributor to cancer-related death throughout the world. Here, we first revealed the role of histone lysine-specific demethylase 5D (KDM5D) in CRC in males. KDM5D expression in tumor and adjacent tissues of male CRC patients was investigated using immunohistochemistry and RT-qPCR, and the correlation between its expression and patients\' prognosis was analyzed. Downregulation of KDM5D in CRC patients was associated with poor prognoses. Overexpression of KDM5D significantly inhibited the growth and metastasis of CRC in vitro and in vivo. The downstream mechanism of KDM5D in CRC was investigated using bioinformatics analysis, and the regulatory relationship was confirmed by ChIP-qPCR and luciferase reporter assays. KDM5D suppressed E2F1 expression by mediating H3K4me3 demethylation. E2F1, highly expressed in CRC, promoted the expression of FKBP4 at the transcriptional level by binding to the FKBP4 promoter. Finally, rescue experiments revealed that overexpression of FKBP4 significantly reversed the inhibitory effect of KDM5D on CRC growth and metastasis. Collectively, KDM5D exerted an anti-tumor and anti-metastatic in CRC through demethylation in E2F1 and suppression of FKBP4 transcription, which might represent a novel target in CRC treatment in male.
    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
    Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of cancer-related mortality worldwide. Methionyl-tRNA synthetase 2 (Mars2) has been suggested as a biomarker indicating poor prognosis of cancers. This study focuses on the function of Mars2 in GC and the responsible molecules. Mars2 was highly expressed in GC patients according to a transcriptome analysis and the data from the public database, and its high expression was confirmed in the acquired GC cell lines. Downregulation of Mars2 significantly weakened the proliferation, resistance to death, migration and invasion of GC cells. The H3K4me3 modification level was increased in the promoter region of Mars2, which was attributed to reduced abundance of lysine demethylase 5D (KDM5D) in the Mars2 promoter. MicroRNA (miR)-4661-5p was identified as an upstream regulator of KDM5D. Downregulation of miR-4661-5p led to an increase in the expression of KDM5D while a decline in the expression of Mars2, which reduced the malignant behaviors of GC cells; however, the malignant behaviors of GC cells was restored after further inhibition of KDM5D. To conclude, this study suggested that increased Mars2 expression upon miR-4661-5p-mediated KDM5D downregulation is correlated with malignant degree of GC cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号