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
    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
    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)

  • 文章类型: Journal Article
    Human neural stem cells (hNSCs) have long been used as an in vitro model to study neurogenesis and as candidates for nervous system therapy. Many parameters have been considered when evaluating the success of transplantation, but sex of donor and recipients is often not discussed. We investigated two commercial NSC lines, the female hNSC-H9 and male hNSC-H14, and we observed faster growth rates in the male cells. At 4 days of differentiation, male cells presented a significant increase in expression of DCX, an immature neuronal marker, while female cells showed a significant increase in RMST, a long noncoding RNA, which is indispensable during neurogenesis. In addition, expression of neural markers MAP2, PSD95, SYP, DCX, and TUJ1 at day 14 of differentiation suggested a similar differentiation potential in both lines. The most significant differences at day 14 of differentiation were the expression levels of RELN, with almost 100-fold difference between the sexes, and MASH1, with more than 1,000-fold increase in male cells. To evaluate whether some of the observed differences may be sex related, we measured the expression of gametologous genes located on the X- and Y-chromosome. Most noticeable was the increase of Y-encoded demethylases KDM6C (UTY) and KDM5D during differentiation of male cells. Our results indicate that attention should be paid to sex when planning neurogenesis and transplantation experiments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Comparative Study
    Sex bias in immune function has been contributed in part to a preponderance of immune system-related genes (ISRG) on the X-chromosome. We verified whether ISRG are more abundant on the X chromosome as compared to autosomal chromosomes and reflected on the impact of our findings.
    Consulting freely accessible databases, we performed a comparative study consisting of three complementary strategies. First, among coding X/Y-linked genes, the abundance of ISRG was compared to the abundance of genes dedicated to other systems. Genes were assigned considering three criteria: disease, tissue expression, and function (DEF approach). In addition, we carried out two genome-wide approaches to compare the contribution of sex and autosomal chromosomes to immune genes defined by an elevated expression in lymphatic tissues (LTEEG approach) or annotation to an immune system process, GO:0002376 (GO approach).
    The X chromosome had less immune genes than the median of the autosomal chromosomes. Among X-linked genes, ISRG ranked fourth after the reproductive and nervous systems and genes dedicated to development, proliferation and apoptosis. On the Y chromosome, ISRG ranked second, and at the pseudoautosomal region (PAR) first. According to studies on the expression of X-linked genes in a variety of (mostly non-lymphatic) tissues, almost two-thirds of ISRG are expressed without sex bias, and the remaining ISRG presented female and male bias with similar frequency. Various epigenetic controllers, X-linked MSL3 and Y-linked KDM5D and UTY, were preferentially expressed in leukocytes and deserve further attention for a possible role in sex biased expression or its neutralisation.
    The X chromosome is not enriched for ISRG, though particular X-linked genes may be responsible for sex differences in certain immune responses. So far, there is insufficient information on sex-biased expression of X/Y-linked ISRG in leukocytes to draw general conclusions on the impact of X/Y-linked ISRG in immune function. More research on the regulation of the expression X-linked genes is required with attention to 1) female and male mechanisms that may either augment or diminish sex biased expression and 2) tissue-specific expression studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    雄性和雌性有相同的遗传密码,但是基因表达谱通常显示出两种性别之间的差异。小鼠胚胎成纤维细胞(MEF)已被用作测试基因功能的有用工具进行实验。它们的特征还在于表达基因如Y连锁Sry或X连锁Hprt的基于性别的差异。然而,没有关于全球基因表达性别差异的报道。这里,使用下一代RNA测序,我们比较了源自两种性别的MEFs的综合转录组。与女性群体相比,男性组上调了27个差异表达基因(DEGs),其中包括男性特异性组蛋白脱甲基酶KDM5D基因,和7个DEG下调。根据搜索ENCODE分析的结果,研究表明,鉴定出的15个基因的表达可能受到H3K4me1或H3K4me3甲基化的调控。有趣的是,我们证明了两种H3K4甲基化都是通过敲低KDM5D诱导的,这导致在男性MEF中发现的八个DEG模式发生变化。总的来说,这些数据不仅表明KDM5D介导的H3K4去甲基化的重要性,涉及男性MEFs的性二态基因表达,但也可以提供有关MEFs用于实验时基因表达的性别依赖性变化的信息。
    Males and females share the same genetic code, but gene expression profile often displays differences between two sexes. Mouse embryonic fibroblasts (MEFs) have been used to experiment as a useful tool to test gene function. They have also been characterized by gender-based differences in expressed genes such as Y-linked Sry or X-linked Hprt. However, there is no report on sex differences in global gene expression. Here, using the next-generation RNA sequencing, we compared the comprehensive transcriptome of MEFs derived from two sexes. In comparison with the female group, the male group up-regulated 27 differentially expressed genes (DEGs), in which a male-specific histone demethylase KDM5D gene is included, and 7 DEGs were down-regulated. Based on the results by searching the ENCODE analysis, it was shown that the expression of 15 genes identified is potentially regulated by the methylation of H3K4me1 or H3K4me3. Interestingly, we demonstrated that both of H3K4 methylation are induced by knocking down KDM5D, which causes changes in patterns of eight DEGs found in male MEFs. Collectively, these data not only suggest an importance of KDM5D-mediated demethylation of H3K4 involved in the sexually dimorphic gene expression in male MEFs, but also may provide information regarding sex-dependent changes in gene expression when MEFs are used for experiments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The androgen receptor (AR) plays an essential role in prostate cancer, and suppression of its signaling with androgen deprivation therapy (ADT) has been the mainstay of treatment for metastatic hormone-sensitive prostate cancer for more than 70 y. Chemotherapy has been reserved for metastatic castration-resistant prostate cancer (mCRPC). The Eastern Cooperative Oncology Group-led trial E3805: ChemoHormonal Therapy Versus Androgen Ablation Randomized Trial for Extensive Disease in Prostate Cancer (CHAARTED) showed that the addition of docetaxel to ADT prolonged overall survival compared with ADT alone in patients with metastatic hormone-sensitive prostate cancer. This finding suggests that there is an interaction between AR signaling activity and docetaxel sensitivity. Here we demonstrate that the prostate cancer cell lines LNCaP and LAPC4 display markedly different sensitivity to docetaxel with AR activation, and RNA-seq analysis of these cell lines identified KDM5D (lysine-specific demethylase 5D) encoded on the Y chromosome as a potential mediator of this sensitivity. Knocking down KDM5D expression in LNCaP leads to docetaxel resistance in the presence of dihydrotestosterone. KDM5D physically interacts with AR in the nucleus, and regulates its transcriptional activity by demethylating H3K4me3 active transcriptional marks. Attenuating KDM5D expression dysregulates AR signaling, resulting in docetaxel insensitivity. KDM5D deletion was also observed in the LNCaP-derived CRPC cell line 104R2, which displayed docetaxel insensitivity with AR activation, unlike parental LNCaP. Dataset analysis from the Oncomine database revealed significantly decreased KDM5D expression in CRPC and poorer prognosis with low KDM5D expression. Taking these data together, this work indicates that KDM5D modulates the AR axis and that this is associated with altered docetaxel sensitivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号