肠道微生物群衍生代谢物对癌症的影响及其在肿瘤免疫疗法中的潜在应用

Mingyan Zhang, Feng Xie, Fangfang Zhou
{"title":"肠道微生物群衍生代谢物对癌症的影响及其在肿瘤免疫疗法中的潜在应用","authors":"Mingyan Zhang,&nbsp;Feng Xie,&nbsp;Fangfang Zhou","doi":"10.1002/mef2.92","DOIUrl":null,"url":null,"abstract":"<p>In recent publications,<span><sup>1, 2</sup></span> the group of Professor Zhu Shu/Pan Wen and the research team of Wang Liangjing/Chen Shujie, respectively investigated two intestinal microbial metabolites: deoxycholic acid (DCA) and indole-3-propionic acid (IPA). DCA acts on the calcium ion channel plasma membrane Ca<sup>2+</sup> ATPase (PMCA), inhibits the effector function of CD8<sup>+</sup> T cells and consequently promotes colorectal cancer (CRC) growth. IPA activates precursor-exhausted T (T<sub>pex</sub>) cells and induces their transformation into effector T (T<sub>eff</sub>) cells, thereby increasing T-cell infiltration into the tumor tissue and enhancing the efficacy of immunotherapy (Figure 1).</p><p>Zhu et al. screened 73 small-molecule compounds derived from microbial sources to examine their effects on CD8<sup>+</sup> T-cell-mediated cytotoxicity or IFN-γ production, and identified DCA. Subsequent experiments using sodium dodecyl sulfate (SDS) or other chemical inhibitors confirmed that DCA does not lead to cell death. In tumor immunity, CD8<sup>+</sup> T cells are crucial in resisting tumors, with cytokines such as IFN-γ and TNF-α playing key roles in killing tumor cells. CD8<sup>+</sup> T-cell activation relies on Ca<sup>2+</sup> as a second messenger and the cytoplasmic Ca<sup>2+</sup> concentration directly affects CD8<sup>+</sup> T-cell activation. The authors found that DCA could inhibit cytoplasmic Ca<sup>2+</sup> accumulation by monitoring the real-time fluorescence intensity of cytoplasmic Ca<sup>2+</sup> in anti-CD3/CD28-activated CD8<sup>+</sup> T cells.</p><p>NFAT2 is an important transcription factor regulating CD8<sup>+</sup> T cells.<span><sup>3</sup></span> According to immunoblotting results, DCA reduced the transcription of this factor. Using NFAT2-luciferase reporter gene assays, the authors observed that PMA/ionomycin stimulation increased the transcriptional activity of NFAT2. Notably, DCA supplementation attenuated this transcriptional activation, indicating that DCA could inhibit the NFAT2-mediated transcription induced by Ca<sup>2+</sup> influx. To further study the specific effects of DCA, the authors examined several key observations. First, DCA did not reduce the levels of free calcium ions in cell-free culture medium, indicating that DCA does not chelate calcium ions. Second, the calcium-release activated channel (CRAC) was the main inward pathway. The immunosuppressive impact of DCA on CD8<sup>+</sup> T cells persisted despite the presence of the selective CRAC channel blocker, BTP2, indicating that DCA did not affect calcium influx. Third, PMA/ionomycin induces Ca<sup>2+</sup> to be released directly from the endoplasmic reticulum without relying on transporter proteins.<span><sup>1, 4</sup></span> However, DCA inhibits Ca<sup>2+</sup> accumulation, indicating that Ca<sup>2+</sup> may flow out of the cell membrane in large quantities or into intracellular stores. Therefore, having blocked various channels known to suppress cytoplasmic Ca<sup>2+</sup> levels, the authors found that DCA inhibits cytoplasmic Ca<sup>2+</sup> accumulation by enhancing PMCA-mediated Ca<sup>2+</sup> efflux, and validated this using a PMCA inhibitor. However, research into the precise binding site of DCA on PMCA remains limited. Further research is required to elucidate the mechanism by which DCA boosts PMCA activity.</p><p>The authors have proposed a possible therapeutic strategy based on the mechanism of DCA. They isolated a bacteriophage from urban sewage that targets the lysis of <i>C. scindens</i>. This bacteriophage effectively reduces the serum DCA concentration, alleviates tumor growth, and enhances the effector function of CD8<sup>+</sup> T cells, thereby presenting a potential therapeutic avenue for CRC.<span><sup>1</sup></span> Additionally, in previous experiments, the use of the inhibitor LaCl<sub>3</sub>, or interference with shPMCA, inhibited PMCA, similarly alleviating the inhibition of CD8<sup>+</sup> T cells. PMCA inhibitors may be used as an adjuvant therapy for those patients with high DCA-producing bacteria.</p><p>Zhu et al.'s research highlights the enhancement of CD8<sup>+</sup> T cell activity as a pivotal aspect of immunotherapy strategies. Wang et al. found that IPA enhances the activity of CD8<sup>+</sup> T cells. They conducted in-depth research on the mechanism by which IPA improves tumor responsiveness to immune checkpoint blockade (ICB). These studies provide a conceptual basis for an adjunct approach to cancer treatment.</p><p>Wang et al. analyzed microbial community differences by dividing mice into poor-responder group and responder group according to αPD-1 therapy. At the species level, <i>Lactobacillus johnsonii</i> exhibited the largest differences. This is consistent with findings in human CRC tissues, where the abundance of this bacterium was negatively correlated with tumor progression. To further investigate the effects of specific substances, the authors grouped mice and treated them with different culture media containing various bacteria. The results showed that only treatment with <i>L. johnsonii</i>-conditioned medium (Lj.CM) had a good therapeutic effect on tumors, comparable to direct oral administration of <i>L. johnsonii</i>. Moreover, the authors found that the levels of tryptophan-related metabolites, especially IPA, a tryptophan-derived metabolite specific to the intestinal microbiota, were increased in the plasma of mice. Surprisingly, only indole-3-lactate (ILA), a precursor substance that can be converted to IPA, was detected in Lj.CM medium, rather than IPA. Subsequent experiments confirmed that the production of IPA requires the cooperation of <i>L. johnsonii</i> and <i>C. sporogenes</i>.<span><sup>2</sup></span> To determine whether the action of this substance depends on T cells, the authors transferred IPA or <i>L. johnsonii</i> to immunodeficient mice. Both treatments didn't reduce the growth of tumor. Then, IPA-untreated CD8<sup>+</sup> T cells or IPA-pretreated CD8<sup>+</sup> T cells was administered to the mice. The authors found that the CD8<sup>+</sup> T cell group pretreated with IPA showed better tumor suppression compared to the group without IPA, proving that IPA acts through T cells. In subsequent studies using techniques such as single-cell RNA sequencing and single-cell T-cell receptor sequencing, the authors examined the group treated with αPD-1 alone or the IPA group treated with IPA and αPD-1 in combination therapy. The results revealed that IPA diminished naive CD8<sup>+</sup> T cell populations, but augmented the population of T<sub>pex</sub> cells which characterized by high TCF1 expression, and promoted the conversion of T<sub>pex</sub> cells to T<sub>eff</sub> cells, thereby enhancing the responsiveness to ICB.<span><sup>2</sup></span> The authors mapped the results of scRNA-seq to those of scATAC-seq and found that IPA activates T<sub>pex</sub> cells by modifying the H3K27 acetylation of the Tcf7 super-enhancer region.</p><p>Based on these results, the authors simulated the tumor microenvironment in CRC tissues. They found that IPA treatment increased CD8<sup>+</sup> T cell infiltration and TCF-1 expression in tumors, effectively improving the efficacy of αPD-1 immunotherapy. However, more investigations are required to validate the feasibility of employing IPA as an adjunct immunotherapeutic agent.</p><p>In conclusion, two research teams discovered the effects of microbial metabolites in the tumor microenvironment by studying the intestinal microbiota. These two studies provided relevant results from different perspectives on tumor immunity. These discoveries offered potential solutions and research directions for promoting the responsiveness of certain tumors to immunotherapy.</p><p>Mingyan Zhang wrote the manuscript and prepared the figure. Feng Xie provided valuable discussion. Fangfang Zhou approved the final version of the manuscript. All authors have read and approved the final manuscript.</p><p>The authors declare no conflicts of interest.</p><p>The authors have nothing to report.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"3 3","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.92","citationCount":"0","resultStr":"{\"title\":\"The impact of intestinal microbiota-derived metabolites on cancer and their potential application in tumor immunotherapy\",\"authors\":\"Mingyan Zhang,&nbsp;Feng Xie,&nbsp;Fangfang Zhou\",\"doi\":\"10.1002/mef2.92\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>In recent publications,<span><sup>1, 2</sup></span> the group of Professor Zhu Shu/Pan Wen and the research team of Wang Liangjing/Chen Shujie, respectively investigated two intestinal microbial metabolites: deoxycholic acid (DCA) and indole-3-propionic acid (IPA). DCA acts on the calcium ion channel plasma membrane Ca<sup>2+</sup> ATPase (PMCA), inhibits the effector function of CD8<sup>+</sup> T cells and consequently promotes colorectal cancer (CRC) growth. IPA activates precursor-exhausted T (T<sub>pex</sub>) cells and induces their transformation into effector T (T<sub>eff</sub>) cells, thereby increasing T-cell infiltration into the tumor tissue and enhancing the efficacy of immunotherapy (Figure 1).</p><p>Zhu et al. screened 73 small-molecule compounds derived from microbial sources to examine their effects on CD8<sup>+</sup> T-cell-mediated cytotoxicity or IFN-γ production, and identified DCA. Subsequent experiments using sodium dodecyl sulfate (SDS) or other chemical inhibitors confirmed that DCA does not lead to cell death. In tumor immunity, CD8<sup>+</sup> T cells are crucial in resisting tumors, with cytokines such as IFN-γ and TNF-α playing key roles in killing tumor cells. CD8<sup>+</sup> T-cell activation relies on Ca<sup>2+</sup> as a second messenger and the cytoplasmic Ca<sup>2+</sup> concentration directly affects CD8<sup>+</sup> T-cell activation. The authors found that DCA could inhibit cytoplasmic Ca<sup>2+</sup> accumulation by monitoring the real-time fluorescence intensity of cytoplasmic Ca<sup>2+</sup> in anti-CD3/CD28-activated CD8<sup>+</sup> T cells.</p><p>NFAT2 is an important transcription factor regulating CD8<sup>+</sup> T cells.<span><sup>3</sup></span> According to immunoblotting results, DCA reduced the transcription of this factor. Using NFAT2-luciferase reporter gene assays, the authors observed that PMA/ionomycin stimulation increased the transcriptional activity of NFAT2. Notably, DCA supplementation attenuated this transcriptional activation, indicating that DCA could inhibit the NFAT2-mediated transcription induced by Ca<sup>2+</sup> influx. To further study the specific effects of DCA, the authors examined several key observations. First, DCA did not reduce the levels of free calcium ions in cell-free culture medium, indicating that DCA does not chelate calcium ions. Second, the calcium-release activated channel (CRAC) was the main inward pathway. The immunosuppressive impact of DCA on CD8<sup>+</sup> T cells persisted despite the presence of the selective CRAC channel blocker, BTP2, indicating that DCA did not affect calcium influx. Third, PMA/ionomycin induces Ca<sup>2+</sup> to be released directly from the endoplasmic reticulum without relying on transporter proteins.<span><sup>1, 4</sup></span> However, DCA inhibits Ca<sup>2+</sup> accumulation, indicating that Ca<sup>2+</sup> may flow out of the cell membrane in large quantities or into intracellular stores. Therefore, having blocked various channels known to suppress cytoplasmic Ca<sup>2+</sup> levels, the authors found that DCA inhibits cytoplasmic Ca<sup>2+</sup> accumulation by enhancing PMCA-mediated Ca<sup>2+</sup> efflux, and validated this using a PMCA inhibitor. However, research into the precise binding site of DCA on PMCA remains limited. Further research is required to elucidate the mechanism by which DCA boosts PMCA activity.</p><p>The authors have proposed a possible therapeutic strategy based on the mechanism of DCA. They isolated a bacteriophage from urban sewage that targets the lysis of <i>C. scindens</i>. This bacteriophage effectively reduces the serum DCA concentration, alleviates tumor growth, and enhances the effector function of CD8<sup>+</sup> T cells, thereby presenting a potential therapeutic avenue for CRC.<span><sup>1</sup></span> Additionally, in previous experiments, the use of the inhibitor LaCl<sub>3</sub>, or interference with shPMCA, inhibited PMCA, similarly alleviating the inhibition of CD8<sup>+</sup> T cells. PMCA inhibitors may be used as an adjuvant therapy for those patients with high DCA-producing bacteria.</p><p>Zhu et al.'s research highlights the enhancement of CD8<sup>+</sup> T cell activity as a pivotal aspect of immunotherapy strategies. Wang et al. found that IPA enhances the activity of CD8<sup>+</sup> T cells. They conducted in-depth research on the mechanism by which IPA improves tumor responsiveness to immune checkpoint blockade (ICB). These studies provide a conceptual basis for an adjunct approach to cancer treatment.</p><p>Wang et al. analyzed microbial community differences by dividing mice into poor-responder group and responder group according to αPD-1 therapy. At the species level, <i>Lactobacillus johnsonii</i> exhibited the largest differences. This is consistent with findings in human CRC tissues, where the abundance of this bacterium was negatively correlated with tumor progression. To further investigate the effects of specific substances, the authors grouped mice and treated them with different culture media containing various bacteria. The results showed that only treatment with <i>L. johnsonii</i>-conditioned medium (Lj.CM) had a good therapeutic effect on tumors, comparable to direct oral administration of <i>L. johnsonii</i>. Moreover, the authors found that the levels of tryptophan-related metabolites, especially IPA, a tryptophan-derived metabolite specific to the intestinal microbiota, were increased in the plasma of mice. Surprisingly, only indole-3-lactate (ILA), a precursor substance that can be converted to IPA, was detected in Lj.CM medium, rather than IPA. Subsequent experiments confirmed that the production of IPA requires the cooperation of <i>L. johnsonii</i> and <i>C. sporogenes</i>.<span><sup>2</sup></span> To determine whether the action of this substance depends on T cells, the authors transferred IPA or <i>L. johnsonii</i> to immunodeficient mice. Both treatments didn't reduce the growth of tumor. Then, IPA-untreated CD8<sup>+</sup> T cells or IPA-pretreated CD8<sup>+</sup> T cells was administered to the mice. The authors found that the CD8<sup>+</sup> T cell group pretreated with IPA showed better tumor suppression compared to the group without IPA, proving that IPA acts through T cells. In subsequent studies using techniques such as single-cell RNA sequencing and single-cell T-cell receptor sequencing, the authors examined the group treated with αPD-1 alone or the IPA group treated with IPA and αPD-1 in combination therapy. The results revealed that IPA diminished naive CD8<sup>+</sup> T cell populations, but augmented the population of T<sub>pex</sub> cells which characterized by high TCF1 expression, and promoted the conversion of T<sub>pex</sub> cells to T<sub>eff</sub> cells, thereby enhancing the responsiveness to ICB.<span><sup>2</sup></span> The authors mapped the results of scRNA-seq to those of scATAC-seq and found that IPA activates T<sub>pex</sub> cells by modifying the H3K27 acetylation of the Tcf7 super-enhancer region.</p><p>Based on these results, the authors simulated the tumor microenvironment in CRC tissues. They found that IPA treatment increased CD8<sup>+</sup> T cell infiltration and TCF-1 expression in tumors, effectively improving the efficacy of αPD-1 immunotherapy. However, more investigations are required to validate the feasibility of employing IPA as an adjunct immunotherapeutic agent.</p><p>In conclusion, two research teams discovered the effects of microbial metabolites in the tumor microenvironment by studying the intestinal microbiota. These two studies provided relevant results from different perspectives on tumor immunity. These discoveries offered potential solutions and research directions for promoting the responsiveness of certain tumors to immunotherapy.</p><p>Mingyan Zhang wrote the manuscript and prepared the figure. Feng Xie provided valuable discussion. Fangfang Zhou approved the final version of the manuscript. All authors have read and approved the final manuscript.</p><p>The authors declare no conflicts of interest.</p><p>The authors have nothing to report.</p>\",\"PeriodicalId\":74135,\"journal\":{\"name\":\"MedComm - Future medicine\",\"volume\":\"3 3\",\"pages\":\"\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-07-30\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.92\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm - Future medicine\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mef2.92\",\"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 - Future medicine","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mef2.92","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

在最近发表的文章1、2中,朱曙/潘文教授课题组和王良静/陈淑杰研究团队分别研究了两种肠道微生物代谢产物:脱氧胆酸(DCA)和吲哚-3-丙酸(IPA)。DCA作用于钙离子通道质膜Ca2+ ATP酶(PMCA),抑制CD8+ T细胞的效应功能,从而促进结直肠癌(CRC)的生长。IPA能激活前体耗竭的T细胞(Tpex),并诱导它们转化为效应T细胞(Teff),从而增加T细胞对肿瘤组织的浸润,提高免疫疗法的疗效(图1)。随后使用十二烷基硫酸钠(SDS)或其他化学抑制剂进行的实验证实,DCA 不会导致细胞死亡。在肿瘤免疫中,CD8+ T 细胞是抵抗肿瘤的关键,IFN-γ 和 TNF-α 等细胞因子在杀死肿瘤细胞方面发挥着关键作用。CD8+ T细胞的活化依赖于作为第二信使的Ca2+,而细胞质中的Ca2+浓度会直接影响CD8+ T细胞的活化。作者通过监测抗 CD3/CD28 激活的 CD8+ T 细胞中胞质 Ca2+ 的实时荧光强度发现,DCA 可抑制胞质 Ca2+ 的积累。3 根据免疫印迹结果,DCA 降低了该因子的转录。作者使用 NFAT2luciferase 报告基因检测法观察到,PMA/洋霉素刺激增加了 NFAT2 的转录活性。值得注意的是,补充 DCA 可减轻这种转录激活,表明 DCA 可抑制 Ca2+ 流入诱导的 NFAT2 介导的转录。为了进一步研究 DCA 的具体作用,作者研究了几个关键的观察结果。首先,DCA 不会降低无细胞培养基中游离钙离子的水平,这表明 DCA 不会螯合钙离子。其次,钙释放激活通道(CRAC)是主要的内向途径。尽管存在选择性 CRAC 通道阻断剂 BTP2,但 DCA 对 CD8+ T 细胞的免疫抑制作用依然存在,这表明 DCA 并不影响钙离子的流入。第三,PMA/阴离子霉素可诱导 Ca2+ 直接从内质网释放,而不依赖于转运蛋白。因此,在阻断了各种已知可抑制细胞质 Ca2+ 水平的通道后,作者发现 DCA 可通过增强 PMCA 介导的 Ca2+ 外流来抑制细胞质 Ca2+ 的积累,并使用 PMCA 抑制剂验证了这一点。然而,有关 DCA 与 PMCA 精确结合位点的研究仍然有限。作者根据 DCA 的机制提出了一种可能的治疗策略。他们从城市污水中分离出一种噬菌体,这种噬菌体的目标是裂解 C. scindens。这种噬菌体能有效降低血清中的 DCA 浓度,缓解肿瘤生长,增强 CD8+ T 细胞的效应功能,从而为 CRC 提供了一条潜在的治疗途径。1 此外,在之前的实验中,使用抑制剂 LaCl3 或干扰 shPMCA 可抑制 PMCA,同样缓解了对 CD8+ T 细胞的抑制。PMCA抑制剂可作为一种辅助疗法,用于那些DCA分泌细菌较多的患者。Zhu等人的研究强调,增强CD8+ T细胞活性是免疫疗法策略的一个关键方面。Wang 等人发现,IPA 能增强 CD8+ T 细胞的活性。他们对IPA提高肿瘤对免疫检查点阻断(ICB)反应性的机制进行了深入研究。Wang等人根据αPD-1疗法将小鼠分为反应差组和反应好组,分析了微生物群落的差异。在物种水平上,约翰逊乳杆菌的差异最大。这与人类 CRC 组织中的发现一致,在人类 CRC 组织中,这种细菌的丰度与肿瘤进展呈负相关。为了进一步研究特定物质的影响,作者将小鼠分组,用含有各种细菌的不同培养基进行处理。结果显示,只有约翰逊酵母调节培养基(Lj.CM)对肿瘤有良好的治疗效果,与直接口服约翰逊酵母的效果相当。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

The impact of intestinal microbiota-derived metabolites on cancer and their potential application in tumor immunotherapy

The impact of intestinal microbiota-derived metabolites on cancer and their potential application in tumor immunotherapy

In recent publications,1, 2 the group of Professor Zhu Shu/Pan Wen and the research team of Wang Liangjing/Chen Shujie, respectively investigated two intestinal microbial metabolites: deoxycholic acid (DCA) and indole-3-propionic acid (IPA). DCA acts on the calcium ion channel plasma membrane Ca2+ ATPase (PMCA), inhibits the effector function of CD8+ T cells and consequently promotes colorectal cancer (CRC) growth. IPA activates precursor-exhausted T (Tpex) cells and induces their transformation into effector T (Teff) cells, thereby increasing T-cell infiltration into the tumor tissue and enhancing the efficacy of immunotherapy (Figure 1).

Zhu et al. screened 73 small-molecule compounds derived from microbial sources to examine their effects on CD8+ T-cell-mediated cytotoxicity or IFN-γ production, and identified DCA. Subsequent experiments using sodium dodecyl sulfate (SDS) or other chemical inhibitors confirmed that DCA does not lead to cell death. In tumor immunity, CD8+ T cells are crucial in resisting tumors, with cytokines such as IFN-γ and TNF-α playing key roles in killing tumor cells. CD8+ T-cell activation relies on Ca2+ as a second messenger and the cytoplasmic Ca2+ concentration directly affects CD8+ T-cell activation. The authors found that DCA could inhibit cytoplasmic Ca2+ accumulation by monitoring the real-time fluorescence intensity of cytoplasmic Ca2+ in anti-CD3/CD28-activated CD8+ T cells.

NFAT2 is an important transcription factor regulating CD8+ T cells.3 According to immunoblotting results, DCA reduced the transcription of this factor. Using NFAT2-luciferase reporter gene assays, the authors observed that PMA/ionomycin stimulation increased the transcriptional activity of NFAT2. Notably, DCA supplementation attenuated this transcriptional activation, indicating that DCA could inhibit the NFAT2-mediated transcription induced by Ca2+ influx. To further study the specific effects of DCA, the authors examined several key observations. First, DCA did not reduce the levels of free calcium ions in cell-free culture medium, indicating that DCA does not chelate calcium ions. Second, the calcium-release activated channel (CRAC) was the main inward pathway. The immunosuppressive impact of DCA on CD8+ T cells persisted despite the presence of the selective CRAC channel blocker, BTP2, indicating that DCA did not affect calcium influx. Third, PMA/ionomycin induces Ca2+ to be released directly from the endoplasmic reticulum without relying on transporter proteins.1, 4 However, DCA inhibits Ca2+ accumulation, indicating that Ca2+ may flow out of the cell membrane in large quantities or into intracellular stores. Therefore, having blocked various channels known to suppress cytoplasmic Ca2+ levels, the authors found that DCA inhibits cytoplasmic Ca2+ accumulation by enhancing PMCA-mediated Ca2+ efflux, and validated this using a PMCA inhibitor. However, research into the precise binding site of DCA on PMCA remains limited. Further research is required to elucidate the mechanism by which DCA boosts PMCA activity.

The authors have proposed a possible therapeutic strategy based on the mechanism of DCA. They isolated a bacteriophage from urban sewage that targets the lysis of C. scindens. This bacteriophage effectively reduces the serum DCA concentration, alleviates tumor growth, and enhances the effector function of CD8+ T cells, thereby presenting a potential therapeutic avenue for CRC.1 Additionally, in previous experiments, the use of the inhibitor LaCl3, or interference with shPMCA, inhibited PMCA, similarly alleviating the inhibition of CD8+ T cells. PMCA inhibitors may be used as an adjuvant therapy for those patients with high DCA-producing bacteria.

Zhu et al.'s research highlights the enhancement of CD8+ T cell activity as a pivotal aspect of immunotherapy strategies. Wang et al. found that IPA enhances the activity of CD8+ T cells. They conducted in-depth research on the mechanism by which IPA improves tumor responsiveness to immune checkpoint blockade (ICB). These studies provide a conceptual basis for an adjunct approach to cancer treatment.

Wang et al. analyzed microbial community differences by dividing mice into poor-responder group and responder group according to αPD-1 therapy. At the species level, Lactobacillus johnsonii exhibited the largest differences. This is consistent with findings in human CRC tissues, where the abundance of this bacterium was negatively correlated with tumor progression. To further investigate the effects of specific substances, the authors grouped mice and treated them with different culture media containing various bacteria. The results showed that only treatment with L. johnsonii-conditioned medium (Lj.CM) had a good therapeutic effect on tumors, comparable to direct oral administration of L. johnsonii. Moreover, the authors found that the levels of tryptophan-related metabolites, especially IPA, a tryptophan-derived metabolite specific to the intestinal microbiota, were increased in the plasma of mice. Surprisingly, only indole-3-lactate (ILA), a precursor substance that can be converted to IPA, was detected in Lj.CM medium, rather than IPA. Subsequent experiments confirmed that the production of IPA requires the cooperation of L. johnsonii and C. sporogenes.2 To determine whether the action of this substance depends on T cells, the authors transferred IPA or L. johnsonii to immunodeficient mice. Both treatments didn't reduce the growth of tumor. Then, IPA-untreated CD8+ T cells or IPA-pretreated CD8+ T cells was administered to the mice. The authors found that the CD8+ T cell group pretreated with IPA showed better tumor suppression compared to the group without IPA, proving that IPA acts through T cells. In subsequent studies using techniques such as single-cell RNA sequencing and single-cell T-cell receptor sequencing, the authors examined the group treated with αPD-1 alone or the IPA group treated with IPA and αPD-1 in combination therapy. The results revealed that IPA diminished naive CD8+ T cell populations, but augmented the population of Tpex cells which characterized by high TCF1 expression, and promoted the conversion of Tpex cells to Teff cells, thereby enhancing the responsiveness to ICB.2 The authors mapped the results of scRNA-seq to those of scATAC-seq and found that IPA activates Tpex cells by modifying the H3K27 acetylation of the Tcf7 super-enhancer region.

Based on these results, the authors simulated the tumor microenvironment in CRC tissues. They found that IPA treatment increased CD8+ T cell infiltration and TCF-1 expression in tumors, effectively improving the efficacy of αPD-1 immunotherapy. However, more investigations are required to validate the feasibility of employing IPA as an adjunct immunotherapeutic agent.

In conclusion, two research teams discovered the effects of microbial metabolites in the tumor microenvironment by studying the intestinal microbiota. These two studies provided relevant results from different perspectives on tumor immunity. These discoveries offered potential solutions and research directions for promoting the responsiveness of certain tumors to immunotherapy.

Mingyan Zhang wrote the manuscript and prepared the figure. Feng Xie provided valuable discussion. Fangfang Zhou approved the final version of the manuscript. All authors have read and approved the final manuscript.

The authors declare no conflicts of interest.

The authors have nothing to report.

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
1.00
自引率
0.00%
发文量
0
×
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