肿瘤细胞利用醋酸来促进肿瘤生长和逃避免疫反应

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
MedComm Pub Date : 2024-08-31 DOI:10.1002/mco2.717
Peng Sun, Juanjuan Liu, Deliang Guo
{"title":"肿瘤细胞利用醋酸来促进肿瘤生长和逃避免疫反应","authors":"Peng Sun,&nbsp;Juanjuan Liu,&nbsp;Deliang Guo","doi":"10.1002/mco2.717","DOIUrl":null,"url":null,"abstract":"<p>A recent study from Zhimin Lu's group published in <i>Nature Metabolism</i><span><sup>1</sup></span> demonstrates that acetate reprogrammed cancer cell metabolism and promoted tumor immune evasion. Notably, nutrients, such as glucose and short-chain fatty acids (SCFAs), in the tumor microenvironment impact tumor growth.<span><sup>1</sup></span> It is well known that tumor cells, regardless of oxygen supply, utilize glucose to produce ATP and building blocks for macromolecule synthesis<span><sup>2, 3</sup></span>; however, recent research has shown that glucose uptake by these cells not only supports the Warburg effect but also triggers non-metabolic functions.<span><sup>4</sup></span> Similar to glucose as a nutrient derived from the diet, acetate, as a main SCFA, is also enriched in the tumor microenvironment. Acetate plays a critical role in mitochondrial oxidation, lipogenesis, and histone acetylation to support tumor cell growth.<span><sup>5</sup></span> However, it remains unclear whether acetate contributes to tumor cell proliferation and immune evasion by directly influencing oncogenic proteins.</p><p>Through metabolomic analysis of human non-small cell lung cancer (NSCLC) specimens, Lu's team revealed that acetate was the most abundant short-chain fatty acid (SCFA). They found that the carbon-13 (<sup>13</sup>C)- or deuterium (<sup>3</sup>D)-labeled acetate was more enriched in lung tumor tissues and tumor cells than in normal lung tissues and tumor interstitial fluid in mice, with a corresponding increase in <sup>13</sup>C-acetyl-CoA in tumor tissues.<span><sup>1</sup></span> Depletion studies of monocarboxylate transporters (MCT)1-4 and sodium-coupled MCT (SMCT)1-2 showed that only the depletion of MCT1, which is highly expressed in NSCLC tissues, led to reduced levels of acetate, acetyl-CoA, and synthesized fatty acids in tumor cells or mouse lung tumors. These results indicate that highly expressed MCT1 transports acetate into tumor cells. Notably, acetate supported tumor cell proliferation and mouse tumor growth only under conditions of low glucose or depletion of glucose transporters GLUT1 and GLUT3,<span><sup>1</sup></span> suggesting that glucose is a primary resource for tumor growth and that acetate counteracts energy stress to sustain tumor cell proliferation.</p><p>In addition to supporting anabolic synthesis, acetate-derived acetyl-CoA, produced by the enzyme acetyl-CoA synthetase 2 (ACSS2), is utilized for protein acetylation. Mass spectrometry analysis of cellular immunoprecipitates with an anti-acetylated lysine antibody showed that acetate increased both the acetylation and expression of c-Myc. In addition, acetate increased the interaction between c-Myc and dihydrolipoamide S-acetyltransferase (DLAT), a component of pyruvate dehydrogenase complex (PDC).<span><sup>1</sup></span> Remarkably, purified DLAT was able to acetylate purified c-Myc at the K148 site in vitro. In NSCLC cells, depletion of DLAT reduced the acetylation and expression of c-Myc at K148, while increasing c-Myc polyubiquitylation. Consistently, acetylation-dead c-Myc K148R and acetylation-mimicking c-Myc K148Q mutations decreased and increased c-Myc half-life, respectively.<span><sup>1</sup></span> These results indicate that DLAT acts as a bona fide protein acetyltransferase to acetylate and stabilize c-Myc.</p><p>Through mass spectrometry analyses, ubiquitin-specific peptidase 10 (USP10) was identified as a c-Myc-associated protein and deubiquitylated c-Myc.<span><sup>1</sup></span> A GST pulldown assay demonstrated that DLAT-mediated c-Myc acetylation facilitated the binding of wild-type (WT) c-Myc, but not the c-Myc K148R mutant, to USP10. Acetate supplementation increased the binding of USP10 to c-Myc but not c-Myc K148R in a DLAT expression-dependent manner whereas c-Myc K148Q increased its association with USP10 compared to its WT counterpart.<span><sup>1</sup></span> Additionally, depletion of USP10 increased c-Myc polyubiquitylation and degradation, and these effects were not reversed by reconstituted USP10 expression when DLAT was depleted. These results indicate that acetate-enhanced and DLAT-mediated c-Myc K148 acetylation induces the binding of USP10 to c-Myc, leading to c-Myc deubiquitylation and stabilization.</p><p>c-Myc activation is known to induce expression of <i>CD274</i> (encoding PD-L1), <i>CCND1</i> (encoding cyclin D1), <i>LDHA</i> (encoding lactate dehydrogenase A), and <i>MCT1</i>.<span><sup>1</sup></span> As expected, acetate supplementation under low-glucose conditions enhanced the expression of PD-L1, cyclin D1, LDHA, and MCT1, MCT1-dependent acetate uptake, lactate production, and tumor cell proliferation.<span><sup>1</sup></span> In addition, coculture of ovalbumin (OVA)-expressing mouse lung cancer cells with mouse CD8<sup>+</sup> T cells expressing a transgenic T cell receptor (TCR) specific for an ovalbumin peptide showed that the acetate-treated tumor cells inhibited the expression of interleukin-2 and interferon-γ in CD8<sup>+</sup> T cells. These acetate-induced effects were abolished by the depletion of MCT1, ACSS2, DLAT, and USP10, or by the knockin expression of the c-Myc K148R mutant.<span><sup>1</sup></span> These results indicate that acetate facilitates acetate uptake, glycolysis, and NSCLC cell proliferation, and PD-L1-dependent inhibition of CD8<sup>+</sup> T-cell activation dependent on the acetate-MCT1-ACSS2-DLAT-USP10-c-Myc axis (Figure 1).</p><p>Analyses of single-cell sequencing datasets from NSCLC and small-cell lung cancer tissues showed that <i>MCT1</i> mRNA levels in tumor cells were higher than those in tumor-infiltrating lymphocytes (TILs).<span><sup>1</sup></span><sup>13</sup>C<sub>2</sub>-acetate isotope-tracing experiments showed that murine lung cancer cells exhibited much higher acetate uptake than tumor-infiltrating CD45<sup>+</sup> leukocytes in mouse lung, suggesting that acetate is more efficiently taken up by tumor cells than TILs.<span><sup>1</sup></span> Time-of-flight mass cytometry (CyTOF) analysis of mouse tumors revealed that acetate supplementation in drinking water decreased the infiltration of cytotoxic CD8<sup>+</sup> T cells, CD4<sup>+</sup> T helper 1 (Th1) cells, and M1 macrophages and increased the infiltration of CD4<sup>+</sup> (Th2) cells and myeloid-derived suppressor cells (MDSCs) in mouse tumors. Acetate also elevated the levels of pro-tumor cytokines, chemokines, and growth factors while decreasing the production of anti-tumor cytokines and factors. This was accompanied by increased tumor tissue expression of c-Myc, Ki-67, MCT1, LDHA, and PD-L1, as well as decreased CD8<sup>+</sup> T-cell infiltration and granzyme B expression. These changes promoted tumor growth and shortened mouse survival time in the presence of glycolysis inhibitor treatment. Reconstituted expression of c-Myc K148R, depletion of MCT1, ACSS2, DLAT and USP10, or treatment with the USP10 inhibitor spautin-1 diminished acetate-induced effect in mice.<span><sup>1</sup></span> Notably, combined treatment with spautin-1 and an anti-PD-1 antibody had an additive effect. These findings indicate that acetate-mediated acetylation of c-Myc at K148 in tumor cells creates an immunosuppressive tumor microenvironment and promotes tumor growth. Inhibition of this pathway eliminates the acetate-induced effect and enhances the efficacy of immune checkpoint blockade therapy. Analyses of 90 human NSCLC tissues showed that c-Myc K148 acetylation levels were positively associated with the MCT1, c-Myc, and PD-L1 expression levels and inversely correlated with CD8<sup>+</sup> T cell infiltration. In addition, c-Myc K148 acetylation or USP10 expression levels were associated with poor survival of the patients.</p><p>Tumor heterogeneity is evident in the differing levels of GLUT1 expression within tumor tissues. This study showed that tumor cells with low GLUT expression or glucose uptake can use acetate as a primary source for acetyl-CoA production and lipid biosynthesis. Acetate also induces c-Myc-dependent MCT1 expression at the transcriptional level. As a result, acetate uptake is further amplified by the acetate-c-Myc-MCT1 positive feedback loop in tumor cells. Importantly, in addition to its role as a metabolic carbon source, acetate reprogrammed tumor cell metabolism and promoted immune evasion through posttranslational modification of c-Myc, which depends on the moonlighting protein acetyltransferase activity of DLAT. Thus, these findings underscore not only the potential of using labeled acetate for cancer diagnosis and monitoring tumor growth but also highlight the possibility of targeting the MCT1-ACSS2-DLAT-USP10-c-Myc signaling axis for cancer treatment, including enhancing the response to immune checkpoint blockade therapy. Importantly, the finding that acetate promotes tumor growth through tumor cell-intrinsic signaling and tumor immune evasion through the regulation of immune checkpoints highlights the significant impact of dietary components on cancer progression, revealing the potential of dietary-based therapy for cancer treatment.</p><p>Deliang Guo, Peng Sun, and Juanjuan Liu conceptualized the writing. All authors together wrote the manuscript. All authors have read and approved the article.</p><p>The authors declare no conflict of interest.</p><p>N/A</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":null,"pages":null},"PeriodicalIF":10.7000,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.717","citationCount":"0","resultStr":"{\"title\":\"Tumor cells utilize acetate for tumor growth and immune evasion\",\"authors\":\"Peng Sun,&nbsp;Juanjuan Liu,&nbsp;Deliang Guo\",\"doi\":\"10.1002/mco2.717\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>A recent study from Zhimin Lu's group published in <i>Nature Metabolism</i><span><sup>1</sup></span> demonstrates that acetate reprogrammed cancer cell metabolism and promoted tumor immune evasion. Notably, nutrients, such as glucose and short-chain fatty acids (SCFAs), in the tumor microenvironment impact tumor growth.<span><sup>1</sup></span> It is well known that tumor cells, regardless of oxygen supply, utilize glucose to produce ATP and building blocks for macromolecule synthesis<span><sup>2, 3</sup></span>; however, recent research has shown that glucose uptake by these cells not only supports the Warburg effect but also triggers non-metabolic functions.<span><sup>4</sup></span> Similar to glucose as a nutrient derived from the diet, acetate, as a main SCFA, is also enriched in the tumor microenvironment. Acetate plays a critical role in mitochondrial oxidation, lipogenesis, and histone acetylation to support tumor cell growth.<span><sup>5</sup></span> However, it remains unclear whether acetate contributes to tumor cell proliferation and immune evasion by directly influencing oncogenic proteins.</p><p>Through metabolomic analysis of human non-small cell lung cancer (NSCLC) specimens, Lu's team revealed that acetate was the most abundant short-chain fatty acid (SCFA). They found that the carbon-13 (<sup>13</sup>C)- or deuterium (<sup>3</sup>D)-labeled acetate was more enriched in lung tumor tissues and tumor cells than in normal lung tissues and tumor interstitial fluid in mice, with a corresponding increase in <sup>13</sup>C-acetyl-CoA in tumor tissues.<span><sup>1</sup></span> Depletion studies of monocarboxylate transporters (MCT)1-4 and sodium-coupled MCT (SMCT)1-2 showed that only the depletion of MCT1, which is highly expressed in NSCLC tissues, led to reduced levels of acetate, acetyl-CoA, and synthesized fatty acids in tumor cells or mouse lung tumors. These results indicate that highly expressed MCT1 transports acetate into tumor cells. Notably, acetate supported tumor cell proliferation and mouse tumor growth only under conditions of low glucose or depletion of glucose transporters GLUT1 and GLUT3,<span><sup>1</sup></span> suggesting that glucose is a primary resource for tumor growth and that acetate counteracts energy stress to sustain tumor cell proliferation.</p><p>In addition to supporting anabolic synthesis, acetate-derived acetyl-CoA, produced by the enzyme acetyl-CoA synthetase 2 (ACSS2), is utilized for protein acetylation. Mass spectrometry analysis of cellular immunoprecipitates with an anti-acetylated lysine antibody showed that acetate increased both the acetylation and expression of c-Myc. In addition, acetate increased the interaction between c-Myc and dihydrolipoamide S-acetyltransferase (DLAT), a component of pyruvate dehydrogenase complex (PDC).<span><sup>1</sup></span> Remarkably, purified DLAT was able to acetylate purified c-Myc at the K148 site in vitro. In NSCLC cells, depletion of DLAT reduced the acetylation and expression of c-Myc at K148, while increasing c-Myc polyubiquitylation. Consistently, acetylation-dead c-Myc K148R and acetylation-mimicking c-Myc K148Q mutations decreased and increased c-Myc half-life, respectively.<span><sup>1</sup></span> These results indicate that DLAT acts as a bona fide protein acetyltransferase to acetylate and stabilize c-Myc.</p><p>Through mass spectrometry analyses, ubiquitin-specific peptidase 10 (USP10) was identified as a c-Myc-associated protein and deubiquitylated c-Myc.<span><sup>1</sup></span> A GST pulldown assay demonstrated that DLAT-mediated c-Myc acetylation facilitated the binding of wild-type (WT) c-Myc, but not the c-Myc K148R mutant, to USP10. Acetate supplementation increased the binding of USP10 to c-Myc but not c-Myc K148R in a DLAT expression-dependent manner whereas c-Myc K148Q increased its association with USP10 compared to its WT counterpart.<span><sup>1</sup></span> Additionally, depletion of USP10 increased c-Myc polyubiquitylation and degradation, and these effects were not reversed by reconstituted USP10 expression when DLAT was depleted. These results indicate that acetate-enhanced and DLAT-mediated c-Myc K148 acetylation induces the binding of USP10 to c-Myc, leading to c-Myc deubiquitylation and stabilization.</p><p>c-Myc activation is known to induce expression of <i>CD274</i> (encoding PD-L1), <i>CCND1</i> (encoding cyclin D1), <i>LDHA</i> (encoding lactate dehydrogenase A), and <i>MCT1</i>.<span><sup>1</sup></span> As expected, acetate supplementation under low-glucose conditions enhanced the expression of PD-L1, cyclin D1, LDHA, and MCT1, MCT1-dependent acetate uptake, lactate production, and tumor cell proliferation.<span><sup>1</sup></span> In addition, coculture of ovalbumin (OVA)-expressing mouse lung cancer cells with mouse CD8<sup>+</sup> T cells expressing a transgenic T cell receptor (TCR) specific for an ovalbumin peptide showed that the acetate-treated tumor cells inhibited the expression of interleukin-2 and interferon-γ in CD8<sup>+</sup> T cells. These acetate-induced effects were abolished by the depletion of MCT1, ACSS2, DLAT, and USP10, or by the knockin expression of the c-Myc K148R mutant.<span><sup>1</sup></span> These results indicate that acetate facilitates acetate uptake, glycolysis, and NSCLC cell proliferation, and PD-L1-dependent inhibition of CD8<sup>+</sup> T-cell activation dependent on the acetate-MCT1-ACSS2-DLAT-USP10-c-Myc axis (Figure 1).</p><p>Analyses of single-cell sequencing datasets from NSCLC and small-cell lung cancer tissues showed that <i>MCT1</i> mRNA levels in tumor cells were higher than those in tumor-infiltrating lymphocytes (TILs).<span><sup>1</sup></span><sup>13</sup>C<sub>2</sub>-acetate isotope-tracing experiments showed that murine lung cancer cells exhibited much higher acetate uptake than tumor-infiltrating CD45<sup>+</sup> leukocytes in mouse lung, suggesting that acetate is more efficiently taken up by tumor cells than TILs.<span><sup>1</sup></span> Time-of-flight mass cytometry (CyTOF) analysis of mouse tumors revealed that acetate supplementation in drinking water decreased the infiltration of cytotoxic CD8<sup>+</sup> T cells, CD4<sup>+</sup> T helper 1 (Th1) cells, and M1 macrophages and increased the infiltration of CD4<sup>+</sup> (Th2) cells and myeloid-derived suppressor cells (MDSCs) in mouse tumors. Acetate also elevated the levels of pro-tumor cytokines, chemokines, and growth factors while decreasing the production of anti-tumor cytokines and factors. This was accompanied by increased tumor tissue expression of c-Myc, Ki-67, MCT1, LDHA, and PD-L1, as well as decreased CD8<sup>+</sup> T-cell infiltration and granzyme B expression. These changes promoted tumor growth and shortened mouse survival time in the presence of glycolysis inhibitor treatment. Reconstituted expression of c-Myc K148R, depletion of MCT1, ACSS2, DLAT and USP10, or treatment with the USP10 inhibitor spautin-1 diminished acetate-induced effect in mice.<span><sup>1</sup></span> Notably, combined treatment with spautin-1 and an anti-PD-1 antibody had an additive effect. These findings indicate that acetate-mediated acetylation of c-Myc at K148 in tumor cells creates an immunosuppressive tumor microenvironment and promotes tumor growth. Inhibition of this pathway eliminates the acetate-induced effect and enhances the efficacy of immune checkpoint blockade therapy. Analyses of 90 human NSCLC tissues showed that c-Myc K148 acetylation levels were positively associated with the MCT1, c-Myc, and PD-L1 expression levels and inversely correlated with CD8<sup>+</sup> T cell infiltration. In addition, c-Myc K148 acetylation or USP10 expression levels were associated with poor survival of the patients.</p><p>Tumor heterogeneity is evident in the differing levels of GLUT1 expression within tumor tissues. This study showed that tumor cells with low GLUT expression or glucose uptake can use acetate as a primary source for acetyl-CoA production and lipid biosynthesis. Acetate also induces c-Myc-dependent MCT1 expression at the transcriptional level. As a result, acetate uptake is further amplified by the acetate-c-Myc-MCT1 positive feedback loop in tumor cells. Importantly, in addition to its role as a metabolic carbon source, acetate reprogrammed tumor cell metabolism and promoted immune evasion through posttranslational modification of c-Myc, which depends on the moonlighting protein acetyltransferase activity of DLAT. Thus, these findings underscore not only the potential of using labeled acetate for cancer diagnosis and monitoring tumor growth but also highlight the possibility of targeting the MCT1-ACSS2-DLAT-USP10-c-Myc signaling axis for cancer treatment, including enhancing the response to immune checkpoint blockade therapy. Importantly, the finding that acetate promotes tumor growth through tumor cell-intrinsic signaling and tumor immune evasion through the regulation of immune checkpoints highlights the significant impact of dietary components on cancer progression, revealing the potential of dietary-based therapy for cancer treatment.</p><p>Deliang Guo, Peng Sun, and Juanjuan Liu conceptualized the writing. All authors together wrote the manuscript. All authors have read and approved the article.</p><p>The authors declare no conflict of interest.</p><p>N/A</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2024-08-31\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.717\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.717\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.717","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

1 这些结果表明,醋酸促进醋酸摄取、糖酵解和 NSCLC 细胞增殖,以及依赖醋酸-MCT1-ACSS2-DLAT-USP10-c-Myc 轴的 PD-L1 依赖性抑制 CD8+ T 细胞活化(图 1)。对 NSCLC 和小细胞肺癌组织单细胞测序数据集的分析表明,肿瘤细胞中的 MCT1 mRNA 水平高于肿瘤浸润淋巴细胞(TILs)。113C2-乙酸酯同位素示踪实验表明,小鼠肺癌细胞对乙酸酯的吸收率远高于小鼠肺部的肿瘤浸润CD45+白细胞,这表明肿瘤细胞对乙酸酯的吸收率高于TILs。对小鼠肿瘤进行的飞行时间质量细胞计数法(CyTOF)分析表明,在饮用水中添加醋酸盐可减少小鼠肿瘤中细胞毒性 CD8+ T 细胞、CD4+ T 辅助 1(Th1)细胞和 M1 巨噬细胞的浸润,增加 CD4+(Th2)细胞和髓源性抑制细胞(MDSCs)的浸润。醋酸盐还能提高促肿瘤细胞因子、趋化因子和生长因子的水平,同时减少抗肿瘤细胞因子和因子的产生。与此同时,肿瘤组织中 c-Myc、Ki-67、MCT1、LDHA 和 PD-L1 的表达也增加了,CD8+ T 细胞浸润和颗粒酶 B 的表达也减少了。这些变化促进了肿瘤的生长,并缩短了小鼠在糖酵解抑制剂治疗下的存活时间。c-Myc K148R 的重组表达,MCT1、ACSS2、DLAT 和 USP10 的耗竭,或 USP10 抑制剂 spautin-1 的处理都会减弱乙酸盐诱导的小鼠效应1。这些研究结果表明,醋酸介导的肿瘤细胞中 c-Myc 在 K148 处的乙酰化会形成免疫抑制性肿瘤微环境,并促进肿瘤生长。抑制这一通路可消除乙酸盐诱导的效应,提高免疫检查点阻断疗法的疗效。对90例人类NSCLC组织的分析表明,c-Myc K148乙酰化水平与MCT1、c-Myc和PD-L1表达水平呈正相关,与CD8+ T细胞浸润呈反相关。此外,c-Myc K148 乙酰化或 USP10 表达水平与患者生存率低有关。这项研究表明,GLUT 表达或葡萄糖摄取量低的肿瘤细胞可将乙酸作为乙酰-CoA 生成和脂质生物合成的主要来源。醋酸还能在转录水平上诱导依赖于 c-Myc 的 MCT1 表达。因此,在肿瘤细胞中,乙酸酯-c-Myc-MCT1 正反馈环会进一步扩大乙酸酯的吸收。重要的是,除了作为代谢碳源的作用外,乙酸盐还通过对 c-Myc 的翻译后修饰重塑了肿瘤细胞的新陈代谢,并促进了免疫逃避,而这种修饰依赖于 DLAT 的月光蛋白乙酰转移酶活性。因此,这些发现不仅强调了利用标记的醋酸酯诊断癌症和监测肿瘤生长的潜力,还突出了靶向 MCT1-ACSS2-DLAT-USP10-c-Myc 信号轴治疗癌症的可能性,包括增强对免疫检查点阻断疗法的反应。重要的是,醋酸酯通过肿瘤细胞内在信号转导促进肿瘤生长,并通过调节免疫检查点逃避肿瘤免疫,这一发现凸显了膳食成分对癌症进展的重要影响,揭示了基于膳食疗法治疗癌症的潜力。所有作者共同撰写了手稿。所有作者均已阅读并认可该文章。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。

Tumor cells utilize acetate for tumor growth and immune evasion

Tumor cells utilize acetate for tumor growth and immune evasion

A recent study from Zhimin Lu's group published in Nature Metabolism1 demonstrates that acetate reprogrammed cancer cell metabolism and promoted tumor immune evasion. Notably, nutrients, such as glucose and short-chain fatty acids (SCFAs), in the tumor microenvironment impact tumor growth.1 It is well known that tumor cells, regardless of oxygen supply, utilize glucose to produce ATP and building blocks for macromolecule synthesis2, 3; however, recent research has shown that glucose uptake by these cells not only supports the Warburg effect but also triggers non-metabolic functions.4 Similar to glucose as a nutrient derived from the diet, acetate, as a main SCFA, is also enriched in the tumor microenvironment. Acetate plays a critical role in mitochondrial oxidation, lipogenesis, and histone acetylation to support tumor cell growth.5 However, it remains unclear whether acetate contributes to tumor cell proliferation and immune evasion by directly influencing oncogenic proteins.

Through metabolomic analysis of human non-small cell lung cancer (NSCLC) specimens, Lu's team revealed that acetate was the most abundant short-chain fatty acid (SCFA). They found that the carbon-13 (13C)- or deuterium (3D)-labeled acetate was more enriched in lung tumor tissues and tumor cells than in normal lung tissues and tumor interstitial fluid in mice, with a corresponding increase in 13C-acetyl-CoA in tumor tissues.1 Depletion studies of monocarboxylate transporters (MCT)1-4 and sodium-coupled MCT (SMCT)1-2 showed that only the depletion of MCT1, which is highly expressed in NSCLC tissues, led to reduced levels of acetate, acetyl-CoA, and synthesized fatty acids in tumor cells or mouse lung tumors. These results indicate that highly expressed MCT1 transports acetate into tumor cells. Notably, acetate supported tumor cell proliferation and mouse tumor growth only under conditions of low glucose or depletion of glucose transporters GLUT1 and GLUT3,1 suggesting that glucose is a primary resource for tumor growth and that acetate counteracts energy stress to sustain tumor cell proliferation.

In addition to supporting anabolic synthesis, acetate-derived acetyl-CoA, produced by the enzyme acetyl-CoA synthetase 2 (ACSS2), is utilized for protein acetylation. Mass spectrometry analysis of cellular immunoprecipitates with an anti-acetylated lysine antibody showed that acetate increased both the acetylation and expression of c-Myc. In addition, acetate increased the interaction between c-Myc and dihydrolipoamide S-acetyltransferase (DLAT), a component of pyruvate dehydrogenase complex (PDC).1 Remarkably, purified DLAT was able to acetylate purified c-Myc at the K148 site in vitro. In NSCLC cells, depletion of DLAT reduced the acetylation and expression of c-Myc at K148, while increasing c-Myc polyubiquitylation. Consistently, acetylation-dead c-Myc K148R and acetylation-mimicking c-Myc K148Q mutations decreased and increased c-Myc half-life, respectively.1 These results indicate that DLAT acts as a bona fide protein acetyltransferase to acetylate and stabilize c-Myc.

Through mass spectrometry analyses, ubiquitin-specific peptidase 10 (USP10) was identified as a c-Myc-associated protein and deubiquitylated c-Myc.1 A GST pulldown assay demonstrated that DLAT-mediated c-Myc acetylation facilitated the binding of wild-type (WT) c-Myc, but not the c-Myc K148R mutant, to USP10. Acetate supplementation increased the binding of USP10 to c-Myc but not c-Myc K148R in a DLAT expression-dependent manner whereas c-Myc K148Q increased its association with USP10 compared to its WT counterpart.1 Additionally, depletion of USP10 increased c-Myc polyubiquitylation and degradation, and these effects were not reversed by reconstituted USP10 expression when DLAT was depleted. These results indicate that acetate-enhanced and DLAT-mediated c-Myc K148 acetylation induces the binding of USP10 to c-Myc, leading to c-Myc deubiquitylation and stabilization.

c-Myc activation is known to induce expression of CD274 (encoding PD-L1), CCND1 (encoding cyclin D1), LDHA (encoding lactate dehydrogenase A), and MCT1.1 As expected, acetate supplementation under low-glucose conditions enhanced the expression of PD-L1, cyclin D1, LDHA, and MCT1, MCT1-dependent acetate uptake, lactate production, and tumor cell proliferation.1 In addition, coculture of ovalbumin (OVA)-expressing mouse lung cancer cells with mouse CD8+ T cells expressing a transgenic T cell receptor (TCR) specific for an ovalbumin peptide showed that the acetate-treated tumor cells inhibited the expression of interleukin-2 and interferon-γ in CD8+ T cells. These acetate-induced effects were abolished by the depletion of MCT1, ACSS2, DLAT, and USP10, or by the knockin expression of the c-Myc K148R mutant.1 These results indicate that acetate facilitates acetate uptake, glycolysis, and NSCLC cell proliferation, and PD-L1-dependent inhibition of CD8+ T-cell activation dependent on the acetate-MCT1-ACSS2-DLAT-USP10-c-Myc axis (Figure 1).

Analyses of single-cell sequencing datasets from NSCLC and small-cell lung cancer tissues showed that MCT1 mRNA levels in tumor cells were higher than those in tumor-infiltrating lymphocytes (TILs).113C2-acetate isotope-tracing experiments showed that murine lung cancer cells exhibited much higher acetate uptake than tumor-infiltrating CD45+ leukocytes in mouse lung, suggesting that acetate is more efficiently taken up by tumor cells than TILs.1 Time-of-flight mass cytometry (CyTOF) analysis of mouse tumors revealed that acetate supplementation in drinking water decreased the infiltration of cytotoxic CD8+ T cells, CD4+ T helper 1 (Th1) cells, and M1 macrophages and increased the infiltration of CD4+ (Th2) cells and myeloid-derived suppressor cells (MDSCs) in mouse tumors. Acetate also elevated the levels of pro-tumor cytokines, chemokines, and growth factors while decreasing the production of anti-tumor cytokines and factors. This was accompanied by increased tumor tissue expression of c-Myc, Ki-67, MCT1, LDHA, and PD-L1, as well as decreased CD8+ T-cell infiltration and granzyme B expression. These changes promoted tumor growth and shortened mouse survival time in the presence of glycolysis inhibitor treatment. Reconstituted expression of c-Myc K148R, depletion of MCT1, ACSS2, DLAT and USP10, or treatment with the USP10 inhibitor spautin-1 diminished acetate-induced effect in mice.1 Notably, combined treatment with spautin-1 and an anti-PD-1 antibody had an additive effect. These findings indicate that acetate-mediated acetylation of c-Myc at K148 in tumor cells creates an immunosuppressive tumor microenvironment and promotes tumor growth. Inhibition of this pathway eliminates the acetate-induced effect and enhances the efficacy of immune checkpoint blockade therapy. Analyses of 90 human NSCLC tissues showed that c-Myc K148 acetylation levels were positively associated with the MCT1, c-Myc, and PD-L1 expression levels and inversely correlated with CD8+ T cell infiltration. In addition, c-Myc K148 acetylation or USP10 expression levels were associated with poor survival of the patients.

Tumor heterogeneity is evident in the differing levels of GLUT1 expression within tumor tissues. This study showed that tumor cells with low GLUT expression or glucose uptake can use acetate as a primary source for acetyl-CoA production and lipid biosynthesis. Acetate also induces c-Myc-dependent MCT1 expression at the transcriptional level. As a result, acetate uptake is further amplified by the acetate-c-Myc-MCT1 positive feedback loop in tumor cells. Importantly, in addition to its role as a metabolic carbon source, acetate reprogrammed tumor cell metabolism and promoted immune evasion through posttranslational modification of c-Myc, which depends on the moonlighting protein acetyltransferase activity of DLAT. Thus, these findings underscore not only the potential of using labeled acetate for cancer diagnosis and monitoring tumor growth but also highlight the possibility of targeting the MCT1-ACSS2-DLAT-USP10-c-Myc signaling axis for cancer treatment, including enhancing the response to immune checkpoint blockade therapy. Importantly, the finding that acetate promotes tumor growth through tumor cell-intrinsic signaling and tumor immune evasion through the regulation of immune checkpoints highlights the significant impact of dietary components on cancer progression, revealing the potential of dietary-based therapy for cancer treatment.

Deliang Guo, Peng Sun, and Juanjuan Liu conceptualized the writing. All authors together wrote the manuscript. All authors have read and approved the article.

The authors declare no conflict of interest.

N/A

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
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
×
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:481959085
Book学术官方微信