WTAP's Dual Role in Disease: Orchestrating Inflammation in Rheumatoid Arthritis and Challenging Renal Clear Cell Carcinoma Outcomes

IF 2.4 4区 医学 Q2 RHEUMATOLOGY
Chen-Yueh Wen, Po-Hung Chen, Chien-Wei Huang, Yen Chin, Chia-Jung Li
{"title":"WTAP's Dual Role in Disease: Orchestrating Inflammation in Rheumatoid Arthritis and Challenging Renal Clear Cell Carcinoma Outcomes","authors":"Chen-Yueh Wen,&nbsp;Po-Hung Chen,&nbsp;Chien-Wei Huang,&nbsp;Yen Chin,&nbsp;Chia-Jung Li","doi":"10.1111/1756-185X.70248","DOIUrl":null,"url":null,"abstract":"<p>The study by Wang et al. titled “WTAP-Mediated m6A Modification of TRAIL-DR4 Suppresses MH7A Cell Apoptosis” highlights the critical role of Wilms tumor 1-associating protein (WTAP), a regulatory component of the N6-methyladenosine (m6A) methyltransferase complex, forms a functional unit with METTL3 and METTL14 to mediate m6A deposition [<span>1</span>]. WTAP promotes mRNA destabilization of TNF-related apoptosis-inducing ligand death receptor 4 (TRAIL-DR4), thereby inhibiting apoptosis and contributing to synovial hyperplasia and persistent inflammation. These findings are consistent with earlier reports demonstrating WTAP's role in inflammatory regulation through m6A-dependent mechanisms, including modulation of cytokine expression in activated macrophages [<span>2</span>]. Building on these foundations, our current investigation expands the scope of WTAP's functional landscape by exploring its relationship with the tumor immune microenvironment, in parallel with its role in inflammatory signaling. This integrated approach is crucial, given the mechanistic overlaps between chronic inflammation and tumorigenesis [<span>3</span>].</p><p>Our study utilized a comprehensive multi-omics approach to elucidate the impact of WTAP expression on genetic landscapes, biological processes, and drug responses in Kidney Renal Clear Cell Carcinoma (KIRC), leveraging data from the TCGA database. We extracted WTAP expression profiles from KIRC patients and visualized the top 20 affected genes using a waterfall plot (Figure 1A). The genetic alterations included oncogenes, tumor suppressor genes, splice site mutations, and frameshift variants, providing a broad genomic context. Comparative analysis of mRNA (Figure 1B) and protein (Figure 1C) expression levels between normal and KIRC tissues revealed a significant reduction in WTAP expression in tumor samples, consistent across TCGA and proteomic datasets.</p><p>Further prognostic analysis using Kaplan–Meier curves demonstrated that low WTAP expression in KIRC is associated with poor overall survival and progression-free survival (Figure 1D,E). This suggests that WTAP's function may be highly tissue- and disease-specific, differing from its anti-apoptotic and anti-inflammatory role in RA. To explore the immunological underpinnings, we assessed T-cell interactions across six independent databases, consistently identifying a positive correlation between WTAP expression and T-cell activation (Figure 1F). We extended our analysis to T-cell-related biological processes, evaluating activation, homeostasis, proliferation, and chemotaxis in KIRC patients stratified by WTAP expression levels (Figure 1G). This contrasts with the MH7A cell model, where WTAP-mediated m6A modification of TRAIL-DR4 inhibits apoptosis and modulates inflammation. Additionally, we interrogated the PRISM Pharmacogenomic Database to assess drug sensitivity and resistance, identifying 20 drugs where high WTAP expression predicted increased sensitivity and 20 where it predicted resistance (Figure 1H). Correlation analyses with IC50 values in the TCGA_KIRC cohort confirmed these trends, with the highly sensitive drug ZK93426 exhibiting negative regulation and procaine showing positive regulation with WTAP levels (Figure 1I). These pharmacogenomic insights suggest that WTAP expression could serve as a biomarker for tailoring therapy in KIRC, contrasting with its therapeutic targeting strategy in the MH7A cell model.</p><p>These findings challenge the uniform application of epigenetic targets like WTAP across malignancies. While the WTAP study demonstrates its role in suppressing apoptosis and inflammation in RA via TRAIL-DR4 mRNA destabilization—confirmed by MeRIP-qPCR and actinomycin D assays [<span>1</span>]—the low WTAP levels in KIRC may reflect a loss of regulatory control over apoptosis and inflammatory pathways, contributing to disease progression. This discrepancy may arise from differences in tumor microenvironment, genetic alterations, or the interplay between m6A modification, immune checkpoints, and inflammation beyond TRAIL-DR4, as seen in other cancers where m6A dysregulation influences tumor immunity [<span>2</span>].</p><p>This editorial advocates for a context-specific approach to targeting epigenetic regulators in oncology, with a particular emphasis on their inflammatory dimensions. The success of WTAP modulation in RA should not be extrapolated to KIRC without further validation. Future research should leverage multi-omics analyses to dissect the pathways linking WTAP, m6A modification, immune responses, and inflammation in KIRC, potentially identifying compensatory mechanisms or alternative targets. Integrating these findings with clinical trials could refine therapeutic strategies, ensuring that WTAP agonists or antagonists are tailored to the specific disease context and its inflammatory profile.</p><p>In conclusion, the low WTAP expression in KIRC, as revealed by our data, contrasts with its protective role in RA and invites a nuanced exploration of its therapeutic potential, particularly in the context of inflammation and the tumor immune microenvironment. Collaborative efforts to bridge epigenetic, immunological, and inflammatory research will be essential to unlocking precision medicine's full potential across diverse cancer landscapes.</p><p>C.-Y.W., P.-H.C., and Y.C. conceived and drafted the manuscript; C.-Y.W. and C.-J.L. provided valuable discussion; C.-J.L. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.</p><p>The authors have nothing to report.</p><p>The authors have nothing to report.</p><p>The authors declare no conflicts of interest.</p>","PeriodicalId":14330,"journal":{"name":"International Journal of Rheumatic Diseases","volume":"28 5","pages":""},"PeriodicalIF":2.4000,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/1756-185X.70248","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"International Journal of Rheumatic Diseases","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/1756-185X.70248","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"RHEUMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

The study by Wang et al. titled “WTAP-Mediated m6A Modification of TRAIL-DR4 Suppresses MH7A Cell Apoptosis” highlights the critical role of Wilms tumor 1-associating protein (WTAP), a regulatory component of the N6-methyladenosine (m6A) methyltransferase complex, forms a functional unit with METTL3 and METTL14 to mediate m6A deposition [1]. WTAP promotes mRNA destabilization of TNF-related apoptosis-inducing ligand death receptor 4 (TRAIL-DR4), thereby inhibiting apoptosis and contributing to synovial hyperplasia and persistent inflammation. These findings are consistent with earlier reports demonstrating WTAP's role in inflammatory regulation through m6A-dependent mechanisms, including modulation of cytokine expression in activated macrophages [2]. Building on these foundations, our current investigation expands the scope of WTAP's functional landscape by exploring its relationship with the tumor immune microenvironment, in parallel with its role in inflammatory signaling. This integrated approach is crucial, given the mechanistic overlaps between chronic inflammation and tumorigenesis [3].

Our study utilized a comprehensive multi-omics approach to elucidate the impact of WTAP expression on genetic landscapes, biological processes, and drug responses in Kidney Renal Clear Cell Carcinoma (KIRC), leveraging data from the TCGA database. We extracted WTAP expression profiles from KIRC patients and visualized the top 20 affected genes using a waterfall plot (Figure 1A). The genetic alterations included oncogenes, tumor suppressor genes, splice site mutations, and frameshift variants, providing a broad genomic context. Comparative analysis of mRNA (Figure 1B) and protein (Figure 1C) expression levels between normal and KIRC tissues revealed a significant reduction in WTAP expression in tumor samples, consistent across TCGA and proteomic datasets.

Further prognostic analysis using Kaplan–Meier curves demonstrated that low WTAP expression in KIRC is associated with poor overall survival and progression-free survival (Figure 1D,E). This suggests that WTAP's function may be highly tissue- and disease-specific, differing from its anti-apoptotic and anti-inflammatory role in RA. To explore the immunological underpinnings, we assessed T-cell interactions across six independent databases, consistently identifying a positive correlation between WTAP expression and T-cell activation (Figure 1F). We extended our analysis to T-cell-related biological processes, evaluating activation, homeostasis, proliferation, and chemotaxis in KIRC patients stratified by WTAP expression levels (Figure 1G). This contrasts with the MH7A cell model, where WTAP-mediated m6A modification of TRAIL-DR4 inhibits apoptosis and modulates inflammation. Additionally, we interrogated the PRISM Pharmacogenomic Database to assess drug sensitivity and resistance, identifying 20 drugs where high WTAP expression predicted increased sensitivity and 20 where it predicted resistance (Figure 1H). Correlation analyses with IC50 values in the TCGA_KIRC cohort confirmed these trends, with the highly sensitive drug ZK93426 exhibiting negative regulation and procaine showing positive regulation with WTAP levels (Figure 1I). These pharmacogenomic insights suggest that WTAP expression could serve as a biomarker for tailoring therapy in KIRC, contrasting with its therapeutic targeting strategy in the MH7A cell model.

These findings challenge the uniform application of epigenetic targets like WTAP across malignancies. While the WTAP study demonstrates its role in suppressing apoptosis and inflammation in RA via TRAIL-DR4 mRNA destabilization—confirmed by MeRIP-qPCR and actinomycin D assays [1]—the low WTAP levels in KIRC may reflect a loss of regulatory control over apoptosis and inflammatory pathways, contributing to disease progression. This discrepancy may arise from differences in tumor microenvironment, genetic alterations, or the interplay between m6A modification, immune checkpoints, and inflammation beyond TRAIL-DR4, as seen in other cancers where m6A dysregulation influences tumor immunity [2].

This editorial advocates for a context-specific approach to targeting epigenetic regulators in oncology, with a particular emphasis on their inflammatory dimensions. The success of WTAP modulation in RA should not be extrapolated to KIRC without further validation. Future research should leverage multi-omics analyses to dissect the pathways linking WTAP, m6A modification, immune responses, and inflammation in KIRC, potentially identifying compensatory mechanisms or alternative targets. Integrating these findings with clinical trials could refine therapeutic strategies, ensuring that WTAP agonists or antagonists are tailored to the specific disease context and its inflammatory profile.

In conclusion, the low WTAP expression in KIRC, as revealed by our data, contrasts with its protective role in RA and invites a nuanced exploration of its therapeutic potential, particularly in the context of inflammation and the tumor immune microenvironment. Collaborative efforts to bridge epigenetic, immunological, and inflammatory research will be essential to unlocking precision medicine's full potential across diverse cancer landscapes.

C.-Y.W., P.-H.C., and Y.C. conceived and drafted the manuscript; C.-Y.W. and C.-J.L. provided valuable discussion; C.-J.L. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

The authors have nothing to report.

The authors have nothing to report.

The authors declare no conflicts of interest.

Abstract Image

WTAP在疾病中的双重作用:类风湿关节炎炎症的协调和肾透明细胞癌结局的挑战
Wang等人的研究题为“WTAP介导的TRAIL-DR4的m6A修饰抑制MH7A细胞凋亡”,强调了Wilms肿瘤1相关蛋白(WTAP)的关键作用,WTAP是n6 -甲基腺苷(m6A)甲基转移酶复合物的调节成分,与METTL3和METTL14形成一个功能单元,介导m6A沉积[1]。WTAP促进tnf相关的凋亡诱导配体死亡受体4 (TRAIL-DR4) mRNA的不稳定,从而抑制细胞凋亡,导致滑膜增生和持续炎症。这些发现与早期的报道一致,证明WTAP通过m6a依赖机制在炎症调节中发挥作用,包括激活巨噬细胞[2]中细胞因子表达的调节。在这些基础上,我们目前的研究通过探索WTAP与肿瘤免疫微环境的关系,以及它在炎症信号传导中的作用,扩大了WTAP功能景观的范围。考虑到慢性炎症和肿瘤发生[3]之间的机制重叠,这种综合方法至关重要。本研究利用TCGA数据库的数据,利用综合多组学方法阐明WTAP表达对肾透明细胞癌(KIRC)遗传景观、生物学过程和药物反应的影响。我们提取了KIRC患者的WTAP表达谱,并使用瀑布图可视化了前20个受影响的基因(图1A)。遗传改变包括致癌基因、肿瘤抑制基因、剪接位点突变和移码变异,提供了广泛的基因组背景。正常组织和KIRC组织之间mRNA(图1B)和蛋白(图1C)表达水平的比较分析显示,肿瘤样本中WTAP表达显著降低,TCGA和蛋白质组学数据集一致。使用Kaplan-Meier曲线的进一步预后分析表明,KIRC中WTAP的低表达与较差的总生存期和无进展生存期相关(图1D,E)。这表明WTAP的功能可能是高度组织和疾病特异性的,不同于它在RA中的抗凋亡和抗炎作用。为了探索免疫学基础,我们在六个独立的数据库中评估了t细胞相互作用,一致地确定了WTAP表达与t细胞激活之间的正相关(图1F)。我们将分析扩展到t细胞相关的生物学过程,评估按WTAP表达水平分层的KIRC患者的激活、稳态、增殖和趋化性(图1G)。这与MH7A细胞模型形成对比,在MH7A细胞模型中,wtap介导的m6A修饰TRAIL-DR4抑制细胞凋亡并调节炎症。此外,我们检索了PRISM药物基因组数据库来评估药物敏感性和耐药性,确定了20种WTAP高表达预测敏感性增加的药物和20种预测耐药性的药物(图1H)。与TCGA_KIRC队列中IC50值的相关分析证实了这些趋势,高敏感药物ZK93426与WTAP水平呈负调节,而普鲁卡因与WTAP水平呈正调节(图1I)。这些药物基因组学见解表明,与MH7A细胞模型中的治疗靶向策略相比,WTAP表达可以作为KIRC中定制治疗的生物标志物。这些发现挑战了表观遗传靶点如WTAP在恶性肿瘤中的统一应用。虽然WTAP研究表明其通过TRAIL-DR4 mRNA不稳定抑制RA细胞凋亡和炎症的作用(MeRIP-qPCR和放线菌素D检测证实了这一点),但KIRC中低WTAP水平可能反映了对细胞凋亡和炎症途径的调节控制缺失,从而导致疾病进展。这种差异可能源于肿瘤微环境的差异、遗传改变或m6A修饰、免疫检查点和TRAIL-DR4以外的炎症之间的相互作用,如在其他m6A失调影响肿瘤免疫的癌症中所见。这篇社论提倡一种针对肿瘤中表观遗传调控因子的具体方法,特别强调它们的炎症维度。在没有进一步验证的情况下,不应将WTAP调制在RA中的成功推断为KIRC。未来的研究应该利用多组学分析来剖析连接WTAP、m6A修饰、免疫反应和KIRC炎症的途径,潜在地确定补偿机制或替代靶点。将这些发现与临床试验相结合,可以改进治疗策略,确保WTAP激动剂或拮抗剂适合特定的疾病背景和炎症特征。 总之,我们的数据显示,KIRC中WTAP的低表达与其在RA中的保护作用形成了对比,并要求对其治疗潜力进行细致的探索,特别是在炎症和肿瘤免疫微环境的背景下。在表观遗传学、免疫学和炎症研究之间建立桥梁的合作努力,对于在不同的癌症领域释放精准医学的全部潜力至关重要。p.h.和Y.C.构思并起草了手稿;c.y.w.和c.j.l.提供了有价值的讨论;c - j - l审阅并编辑了手稿。所有作者都已阅读并同意稿件的出版版本。作者没有什么可报告的。作者没有什么可报告的。作者声明无利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
3.70
自引率
4.00%
发文量
362
审稿时长
1 months
期刊介绍: The International Journal of Rheumatic Diseases (formerly APLAR Journal of Rheumatology) is the official journal of the Asia Pacific League of Associations for Rheumatology. The Journal accepts original articles on clinical or experimental research pertinent to the rheumatic diseases, work on connective tissue diseases and other immune and allergic disorders. The acceptance criteria for all papers are the quality and originality of the research and its significance to our readership. Except where otherwise stated, manuscripts are peer reviewed by two anonymous reviewers and the Editor.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信