KMT2C/D 缺乏通过调节 KDM6A 介导的表观遗传重塑促进乳腺癌转移

Zhao Huang, Wei Zhao
{"title":"KMT2C/D 缺乏通过调节 KDM6A 介导的表观遗传重塑促进乳腺癌转移","authors":"Zhao Huang,&nbsp;Wei Zhao","doi":"10.1002/mog2.95","DOIUrl":null,"url":null,"abstract":"<p>On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (<i>KMT2C)</i> or <i>KMT2D</i> deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.<span><sup>1</sup></span> Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.<span><sup>2</sup></span> TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.<span><sup>3</sup></span></p><p>The most commonly mutated gene in BMs of breast cancer has been reported to be <i>KMT2C/D</i>, and the results of gene sequencing showed that <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of <i>KMT2C/D</i> plays a role in BMs of breast cancer. <i>KMT2C</i> and <i>KMT2D</i> catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), <i>KMT2C</i> and <i>KMT2D</i> profoundly regulate the epigenetic landscape.<span><sup>4</sup></span> However, it is still unclear how <i>KMT2C</i>/<i>D</i> mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.</p><p>Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of <i>KMT2C</i> or <i>KMT2D</i> can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of <i>KMT2C</i> or <i>KMT2D</i> promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.<span><sup>1</sup></span> This study provides not only a new perspective for the treatment of TNBC at the epigenetic level, but also a potential target for the development of targeted therapeutic strategies for BMs.</p><p>First, Seehawer et al. showed that the expression level of <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared with primary breast tumors. To investigate the role of <i>KMT2C/D</i> deletion in BMs, they injected the nonmetastatic mouse breast tumor cell line 168FARN tagged with H2B-mCherry into the mammary fat pad (MFP) of mice and found that while orthotopic tumor growth was unaffected, mCherry+ micrometastatic lesions appeared in the lung, liver, brain in mice injected with <i>KMT2C</i> or <i>KMT2D</i> knockout (KO) 168FARN cells. This suggests that they are indeed derived from KO cells.</p><p>Subsequently, the researchers performed single-cell RNA sequencing (scRNA-seq) on 168FARN-derived primary tumors injected with MFP. Differentially expressed gene analysis found that <i>Ly6a</i>, <i>Bst2</i>, <i>Ifi27l2a</i>, and <i>Stat1</i> expression were significantly upregulated in KO tumor cells, suggesting a proinflammatory environment generated by potential activation of interferon signaling. In addition, they found that the loss of <i>KMT2C</i> and <i>KMT2D</i> had specific effects on different types of cell subsets in the tumor microenvironment.</p><p>To dissect the cell-intrinsic changes caused by the loss of <i>KMT2C</i> or <i>KMT2D</i>, Seehawer et al. focused on histones that are direct targets of <i>KMT2C/D</i>. Using quantitative histone mass spectrometry, immunoblotting and chromatin immunoprecipitation sequencing (ChIP-seq) approaches, they found locus-specific but not global changes of H3K4me1 in both KO cells. By contrast, H3K27me3 was altered globally instead of a locus-specific manner. KDM6A, as an H3K27me3 demethylase, is one of the components of COMPASS complex. KDM6A-ChIP-seq results showed that the peaks in KO cells increased significantly.</p><p>Subsequently, Seehawer et al. performed RNA-seq analysis and found that the loss of <i>KMT2C</i> or <i>KMT2D</i> induced the epithelial-mesenchymal hybriditic transformation (EMT) state, altering the EMT balance and thus promoting metastasis. It is worth mentioning that the role of <i>KMT2C/D</i> deletion in promoting metastasis was observed even in the absence of EMT. Further integrating RNA-seq and ChIP-seq data, they found that <i>KMT2C/D</i> deficiency can regulate gene expression by affecting chromatin state, which may affect tumor metastasis, and MMP3KDM6A may be a mediator of transcriptome changes associated with <i>KMT2C/D</i> deficiency.</p><p>To identify common drivers inducing the epigenetic events and the metastatic phenotype of <i>KMT2C</i>/<i>D</i> KO cells, Seehawer et al. integrated data from ChIP-seq, RNA-seq and scRNA-seq, combined with experimental validation, confirming that <i>MMP3</i> was the only overlapping gene. More importantly, analysis of clinical samples found that <i>MMP3</i> was overexpressed in <i>KMT2C</i>-mutated TNBCs compared with WT tumors, and the frequency of <i>KMT2C</i> mutations was significantly higher in <i>MMP3</i>-high-expressing tumors. Further, downregulation of <i>MMP3</i> by shRNA significantly reduced BMs of <i>KMT2/D</i> KO cells in a mouse model. Existing clinical trials of MMP inhibitors have shown high toxicity and low efficacy. Fortunately, Seehawer et al. found that inhibiting <i>KDM6A</i> by shRNA or pharmacology could reduce the expression of <i>MMP3</i> and inhibit BMs of <i>KMT2C/D</i> KO cells in vivo. In addition, they also found that KDM6A inhibition was tolerable in mice, suggesting that KDM6A inhibitors may have potential clinical application value for TNBC patients (Figure 1).</p><p>Epigenetic regulation is widely considered as a major tumor-suppressive mechanism for many cancer types, including breast cancer.<span><sup>5</sup></span> In summary, this study elucidated the molecular mechanism of chromatin remodeling and histone modification changes caused by <i>KMT2C/D</i> mutations, which indirectly affected the expression of MMP3 through KDM5A, thereby inducing TNBC distant metastasis. Inhibiting the activity of KDM6A can lead to the suppression of MMP3, which represses the metastasis caused by <i>KMT2C/D</i> mutations in TNBC. The results of this study increase the understanding of the mechanism of TNBC BMs, provide a potential target for the development of targeted therapeutic strategies for BMs. As KDM6A inhibitors have not yet been used in clinical treatment for cancers, this study provided valuable information for the development of novel KDM6A-based therapeutic strategies for the treatment of TNBC.</p><p>Wei Zhao provided acquisition, analysis, and interpretation of data. Zhao Huang was responsible for writing, reviewing, and revising the paper. Both authors have read and approved the final manuscript.</p><p>Not applicable.</p><p>Not applicable.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.95","citationCount":"0","resultStr":"{\"title\":\"KMT2C/D deficiency promotes breast cancer metastasis by regulating KDM6A-mediated epigenetic remodeling\",\"authors\":\"Zhao Huang,&nbsp;Wei Zhao\",\"doi\":\"10.1002/mog2.95\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (<i>KMT2C)</i> or <i>KMT2D</i> deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.<span><sup>1</sup></span> Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.<span><sup>2</sup></span> TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.<span><sup>3</sup></span></p><p>The most commonly mutated gene in BMs of breast cancer has been reported to be <i>KMT2C/D</i>, and the results of gene sequencing showed that <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of <i>KMT2C/D</i> plays a role in BMs of breast cancer. <i>KMT2C</i> and <i>KMT2D</i> catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), <i>KMT2C</i> and <i>KMT2D</i> profoundly regulate the epigenetic landscape.<span><sup>4</sup></span> However, it is still unclear how <i>KMT2C</i>/<i>D</i> mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.</p><p>Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of <i>KMT2C</i> or <i>KMT2D</i> can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of <i>KMT2C</i> or <i>KMT2D</i> promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.<span><sup>1</sup></span> This study provides not only a new perspective for the treatment of TNBC at the epigenetic level, but also a potential target for the development of targeted therapeutic strategies for BMs.</p><p>First, Seehawer et al. showed that the expression level of <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared with primary breast tumors. To investigate the role of <i>KMT2C/D</i> deletion in BMs, they injected the nonmetastatic mouse breast tumor cell line 168FARN tagged with H2B-mCherry into the mammary fat pad (MFP) of mice and found that while orthotopic tumor growth was unaffected, mCherry+ micrometastatic lesions appeared in the lung, liver, brain in mice injected with <i>KMT2C</i> or <i>KMT2D</i> knockout (KO) 168FARN cells. This suggests that they are indeed derived from KO cells.</p><p>Subsequently, the researchers performed single-cell RNA sequencing (scRNA-seq) on 168FARN-derived primary tumors injected with MFP. Differentially expressed gene analysis found that <i>Ly6a</i>, <i>Bst2</i>, <i>Ifi27l2a</i>, and <i>Stat1</i> expression were significantly upregulated in KO tumor cells, suggesting a proinflammatory environment generated by potential activation of interferon signaling. In addition, they found that the loss of <i>KMT2C</i> and <i>KMT2D</i> had specific effects on different types of cell subsets in the tumor microenvironment.</p><p>To dissect the cell-intrinsic changes caused by the loss of <i>KMT2C</i> or <i>KMT2D</i>, Seehawer et al. focused on histones that are direct targets of <i>KMT2C/D</i>. Using quantitative histone mass spectrometry, immunoblotting and chromatin immunoprecipitation sequencing (ChIP-seq) approaches, they found locus-specific but not global changes of H3K4me1 in both KO cells. By contrast, H3K27me3 was altered globally instead of a locus-specific manner. KDM6A, as an H3K27me3 demethylase, is one of the components of COMPASS complex. KDM6A-ChIP-seq results showed that the peaks in KO cells increased significantly.</p><p>Subsequently, Seehawer et al. performed RNA-seq analysis and found that the loss of <i>KMT2C</i> or <i>KMT2D</i> induced the epithelial-mesenchymal hybriditic transformation (EMT) state, altering the EMT balance and thus promoting metastasis. It is worth mentioning that the role of <i>KMT2C/D</i> deletion in promoting metastasis was observed even in the absence of EMT. Further integrating RNA-seq and ChIP-seq data, they found that <i>KMT2C/D</i> deficiency can regulate gene expression by affecting chromatin state, which may affect tumor metastasis, and MMP3KDM6A may be a mediator of transcriptome changes associated with <i>KMT2C/D</i> deficiency.</p><p>To identify common drivers inducing the epigenetic events and the metastatic phenotype of <i>KMT2C</i>/<i>D</i> KO cells, Seehawer et al. integrated data from ChIP-seq, RNA-seq and scRNA-seq, combined with experimental validation, confirming that <i>MMP3</i> was the only overlapping gene. More importantly, analysis of clinical samples found that <i>MMP3</i> was overexpressed in <i>KMT2C</i>-mutated TNBCs compared with WT tumors, and the frequency of <i>KMT2C</i> mutations was significantly higher in <i>MMP3</i>-high-expressing tumors. Further, downregulation of <i>MMP3</i> by shRNA significantly reduced BMs of <i>KMT2/D</i> KO cells in a mouse model. Existing clinical trials of MMP inhibitors have shown high toxicity and low efficacy. Fortunately, Seehawer et al. found that inhibiting <i>KDM6A</i> by shRNA or pharmacology could reduce the expression of <i>MMP3</i> and inhibit BMs of <i>KMT2C/D</i> KO cells in vivo. In addition, they also found that KDM6A inhibition was tolerable in mice, suggesting that KDM6A inhibitors may have potential clinical application value for TNBC patients (Figure 1).</p><p>Epigenetic regulation is widely considered as a major tumor-suppressive mechanism for many cancer types, including breast cancer.<span><sup>5</sup></span> In summary, this study elucidated the molecular mechanism of chromatin remodeling and histone modification changes caused by <i>KMT2C/D</i> mutations, which indirectly affected the expression of MMP3 through KDM5A, thereby inducing TNBC distant metastasis. Inhibiting the activity of KDM6A can lead to the suppression of MMP3, which represses the metastasis caused by <i>KMT2C/D</i> mutations in TNBC. The results of this study increase the understanding of the mechanism of TNBC BMs, provide a potential target for the development of targeted therapeutic strategies for BMs. As KDM6A inhibitors have not yet been used in clinical treatment for cancers, this study provided valuable information for the development of novel KDM6A-based therapeutic strategies for the treatment of TNBC.</p><p>Wei Zhao provided acquisition, analysis, and interpretation of data. Zhao Huang was responsible for writing, reviewing, and revising the paper. Both authors have read and approved the final manuscript.</p><p>Not applicable.</p><p>Not applicable.</p>\",\"PeriodicalId\":100902,\"journal\":{\"name\":\"MedComm – Oncology\",\"volume\":\"3 4\",\"pages\":\"\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-10-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.95\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm – Oncology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mog2.95\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm – Oncology","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mog2.95","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

2024年6月26日,Kornelia Polyak团队在《自然-细胞生物学》(Nature Cell Biology)杂志上发表文章,揭示了组蛋白-赖氨酸N-甲基转移酶2 C(KMT2C)或KMT2D缺失以赖氨酸特异性去甲基化酶6 A(KDM6A)依赖方式促进金属蛋白酶3(MMP3)表达,从而驱动三阴性乳腺癌(TNBC)脑转移(BMs)的新机制。1 乳腺癌是最常见的肿瘤,也是全球妇女因肿瘤死亡的主要原因。乳腺癌亚型可根据雌激素受体、孕激素受体和人表皮生长因子受体 2 的表达情况进行分类。2 TNBC 的特点是细胞侵袭性强,可内脏转移至器官,通常是脑、肺和肝,平均生存时间为 18 个月。虽然 TNBC 只占所有乳腺癌的 15%-20%,但却是预后最差的乳腺癌亚型。在免疫疗法出现之前,包括紫杉类、蒽环类和/或铂类在内的全身化疗是 TNBC 的主要一线治疗方案。然而,转移性 TNBC 的中位总生存期仅为 9-12 个月,5 年生存率约为 12%,这是一个严重的未满足的医疗需求。3 有报道称,乳腺癌骨髓瘤中最常见的突变基因是 KMT2C/D,基因测序结果显示,与原发肿瘤相比,KMT2C 和 KMT2D 在远端转移瘤中明显减少,这表明 KMT2C/D 的功能缺失在乳腺癌骨髓瘤中发挥作用。KMT2C 和 KMT2D 可催化未甲基化的 H3K4 位点的甲基化,形成 H3K4me1,这种甲基化可富集在活跃的增强子和启动子区域。这种修饰会招募其他组蛋白修饰酶,包括组蛋白 H3K27 乙酰转移酶(如 P300)和去甲基化酶(如 KDM6A),它们对基因表达的调控至关重要。最近,美国丹娜-法伯癌症研究所的 Marco Seehawer 及其同事发现,在非转移性 TNBC 小鼠模型中,KMT2C 或 KMT2D 的缺失可导致肿瘤转移(尤其是骨髓瘤)。机理研究发现,KMT2C或KMT2D的缺失会以KDM6A依赖的方式促进基质MMP3的表达,从而驱动TNBC的BMs1。这项研究不仅为从表观遗传学水平治疗TNBC提供了新的视角,也为开发针对BMs的靶向治疗策略提供了潜在靶点。为了研究 KMT2C/D 缺失在乳腺肿瘤中的作用,他们将标记有 H2B-mCherry 的非转移性小鼠乳腺肿瘤细胞系 168FARN 注射到小鼠的乳腺脂肪垫(MFP)中,发现虽然正位肿瘤生长不受影响,但注射了 KMT2C 或 KMT2D 基因敲除(KO)168FARN 细胞的小鼠的肺、肝、脑中出现了 mCherry+ 微转移病灶。随后,研究人员对注射了 MFP 的 168FARN 衍生原发肿瘤进行了单细胞 RNA 测序(scRNA-seq)。差异表达基因分析发现,Ly6a、Bst2、Ifi27l2a和Stat1的表达在KO肿瘤细胞中显著上调,这表明干扰素信号的潜在激活产生了一种促炎环境。此外,他们还发现,KMT2C和KMT2D的缺失对肿瘤微环境中不同类型的细胞亚群有特定的影响。为了剖析KMT2C或KMT2D缺失引起的细胞内在变化,Seehawer等人重点研究了作为KMT2C/D直接靶标的组蛋白。利用定量组蛋白质谱、免疫印迹和染色质免疫沉淀测序(ChIP-seq)方法,他们发现在这两种KO细胞中,H3K4me1发生了位点特异性变化,但没有发生全局性变化。相比之下,H3K27me3的变化是全局性的,而不是位点特异性的。KDM6A 是一种 H3K27me3 去甲基化酶,是 COMPASS 复合物的组成成分之一。KDM6A-ChIP-seq结果显示,KO细胞中的峰值显著增加。随后,Seehawer等人进行了RNA-seq分析,发现KMT2C或KMT2D的缺失会诱导上皮-间质杂交转化(EMT)状态,改变EMT平衡,从而促进转移。值得一提的是,即使在没有EMT的情况下,也能观察到KMT2C/D缺失在促进转移方面的作用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
KMT2C/D deficiency promotes breast cancer metastasis by regulating KDM6A-mediated epigenetic remodeling

On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (KMT2C) or KMT2D deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.1 Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.2 TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.3

The most commonly mutated gene in BMs of breast cancer has been reported to be KMT2C/D, and the results of gene sequencing showed that KMT2C and KMT2D were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of KMT2C/D plays a role in BMs of breast cancer. KMT2C and KMT2D catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), KMT2C and KMT2D profoundly regulate the epigenetic landscape.4 However, it is still unclear how KMT2C/D mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.

Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of KMT2C or KMT2D can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of KMT2C or KMT2D promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.1 This study provides not only a new perspective for the treatment of TNBC at the epigenetic level, but also a potential target for the development of targeted therapeutic strategies for BMs.

First, Seehawer et al. showed that the expression level of KMT2C and KMT2D were significantly reduced in distal metastasis compared with primary breast tumors. To investigate the role of KMT2C/D deletion in BMs, they injected the nonmetastatic mouse breast tumor cell line 168FARN tagged with H2B-mCherry into the mammary fat pad (MFP) of mice and found that while orthotopic tumor growth was unaffected, mCherry+ micrometastatic lesions appeared in the lung, liver, brain in mice injected with KMT2C or KMT2D knockout (KO) 168FARN cells. This suggests that they are indeed derived from KO cells.

Subsequently, the researchers performed single-cell RNA sequencing (scRNA-seq) on 168FARN-derived primary tumors injected with MFP. Differentially expressed gene analysis found that Ly6a, Bst2, Ifi27l2a, and Stat1 expression were significantly upregulated in KO tumor cells, suggesting a proinflammatory environment generated by potential activation of interferon signaling. In addition, they found that the loss of KMT2C and KMT2D had specific effects on different types of cell subsets in the tumor microenvironment.

To dissect the cell-intrinsic changes caused by the loss of KMT2C or KMT2D, Seehawer et al. focused on histones that are direct targets of KMT2C/D. Using quantitative histone mass spectrometry, immunoblotting and chromatin immunoprecipitation sequencing (ChIP-seq) approaches, they found locus-specific but not global changes of H3K4me1 in both KO cells. By contrast, H3K27me3 was altered globally instead of a locus-specific manner. KDM6A, as an H3K27me3 demethylase, is one of the components of COMPASS complex. KDM6A-ChIP-seq results showed that the peaks in KO cells increased significantly.

Subsequently, Seehawer et al. performed RNA-seq analysis and found that the loss of KMT2C or KMT2D induced the epithelial-mesenchymal hybriditic transformation (EMT) state, altering the EMT balance and thus promoting metastasis. It is worth mentioning that the role of KMT2C/D deletion in promoting metastasis was observed even in the absence of EMT. Further integrating RNA-seq and ChIP-seq data, they found that KMT2C/D deficiency can regulate gene expression by affecting chromatin state, which may affect tumor metastasis, and MMP3KDM6A may be a mediator of transcriptome changes associated with KMT2C/D deficiency.

To identify common drivers inducing the epigenetic events and the metastatic phenotype of KMT2C/D KO cells, Seehawer et al. integrated data from ChIP-seq, RNA-seq and scRNA-seq, combined with experimental validation, confirming that MMP3 was the only overlapping gene. More importantly, analysis of clinical samples found that MMP3 was overexpressed in KMT2C-mutated TNBCs compared with WT tumors, and the frequency of KMT2C mutations was significantly higher in MMP3-high-expressing tumors. Further, downregulation of MMP3 by shRNA significantly reduced BMs of KMT2/D KO cells in a mouse model. Existing clinical trials of MMP inhibitors have shown high toxicity and low efficacy. Fortunately, Seehawer et al. found that inhibiting KDM6A by shRNA or pharmacology could reduce the expression of MMP3 and inhibit BMs of KMT2C/D KO cells in vivo. In addition, they also found that KDM6A inhibition was tolerable in mice, suggesting that KDM6A inhibitors may have potential clinical application value for TNBC patients (Figure 1).

Epigenetic regulation is widely considered as a major tumor-suppressive mechanism for many cancer types, including breast cancer.5 In summary, this study elucidated the molecular mechanism of chromatin remodeling and histone modification changes caused by KMT2C/D mutations, which indirectly affected the expression of MMP3 through KDM5A, thereby inducing TNBC distant metastasis. Inhibiting the activity of KDM6A can lead to the suppression of MMP3, which represses the metastasis caused by KMT2C/D mutations in TNBC. The results of this study increase the understanding of the mechanism of TNBC BMs, provide a potential target for the development of targeted therapeutic strategies for BMs. As KDM6A inhibitors have not yet been used in clinical treatment for cancers, this study provided valuable information for the development of novel KDM6A-based therapeutic strategies for the treatment of TNBC.

Wei Zhao provided acquisition, analysis, and interpretation of data. Zhao Huang was responsible for writing, reviewing, and revising the paper. Both authors have read and approved the final manuscript.

Not applicable.

Not applicable.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Macrophage-Mediated Myelin Recycling Promotes Malignant Development of Glioblastoma Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention Lomitapide: Targeting METTL3 to Overcome Osimertinib Resistance in NSCLC Through Autophagy Activation Ephrin A1 ligand-based CAR-T cells for immunotherapy of EphA2-positive cancer Analysis of reoperational reason of patients with thyroid cancer and strategies for its diagnosis and treatment: A 6-year single-center retrospective study
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1