首页 > 最新文献

Cancer & Metabolism最新文献

英文 中文
Tamoxifen induces radioresistance through NRF2-mediated metabolic reprogramming in breast cancer. 他莫昔芬通过NRF2介导的乳腺癌代谢重编程诱导放射抗性
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2023-02-08 DOI: 10.1186/s40170-023-00304-4
F V Reinema, F C G J Sweep, G J Adema, W J M Peeters, J W M Martens, J Bussink, P N Span

Background: Recently, we reported that tamoxifen-resistant (TAM-R) breast cancer cells are cross-resistant to irradiation. Here, we investigated the mechanisms associated with tamoxifen-induced radioresistance, aiming to prevent or reverse resistance and improve breast cancer treatment.

Methods: Wild-type ERα-positive MCF7 and ERα-negative MDA-MB-231 breast cancer cells and their TAM-R counterparts were analyzed for cellular metabolism using the Seahorse metabolic analyzer. Real-time ROS production, toxicity, and antioxidant capacity in response to H2O2, tamoxifen, and irradiation were determined. Tumor material from 28 breast cancer patients before and after short-term presurgical tamoxifen (ClinicalTrials.gov Identifier: NCT00738777, August 19, 2008) and cellular material was analyzed for NRF2 gene expression and immunohistochemistry. Re-sensitization of TAM-R cells to irradiation was established using pharmacological inhibition.

Results: TAM-R cells exhibited decreased oxygen consumption and increased glycolysis, suggesting mitochondrial dysfunction. However, this did not explain radioresistance, as cells without mitochondria (Rho-0) were actually more radiosensitive. Real-time measurement of ROS after tamoxifen and H2O2 exposure indicated lower ROS levels and toxicity in TAM-R cells. Consistently, higher antioxidant levels were found in TAM-R cells, providing protection from irradiation-induced ROS. NRF2, a main activator of the antioxidant response, was increased in TAM-R cells and in tumor tissue of patients treated with short-term presurgical tamoxifen. NRF2 inhibition re-sensitized TAM-R cells to irradiation.

Conclusion: Mechanisms underlying tamoxifen-induced radioresistance are linked to cellular adaptations to persistently increased ROS levels, leading to cells with chronically upregulated antioxidant capacity and glycolysis. Pharmacological inhibition of antioxidant responses re-sensitizes breast cancer cells to irradiation.

背景:最近,我们报道了他莫昔芬耐药(TAM-R)乳腺癌细胞对辐照具有交叉耐药性。在此,我们研究了他莫昔芬诱导放射抗性的相关机制,旨在预防或逆转抗性,改善乳腺癌治疗:方法:使用 Seahorse 代谢分析仪分析野生型 ERα 阳性 MCF7 和 ERα 阴性 MDA-MB-231 乳腺癌细胞及其 TAM-R 对应细胞的细胞代谢。还测定了实时 ROS 生成、毒性以及对 H2O2、他莫昔芬和辐照的抗氧化能力。对 28 名乳腺癌患者短期手术前他莫昔芬治疗前后的肿瘤材料(ClinicalTrials.gov Identifier:NCT00738777,2008 年 8 月 19 日)和细胞材料进行了 NRF2 基因表达和免疫组化分析。通过药理抑制作用确定了 TAM-R 细胞对辐照的再敏感性:结果:TAM-R细胞的耗氧量降低,糖酵解增加,表明线粒体功能障碍。然而,这并不能解释放射抗性,因为没有线粒体的细胞(Rho-0)实际上对放射更敏感。他莫昔芬和 H2O2 暴露后的实时 ROS 测量表明,TAM-R 细胞的 ROS 水平和毒性较低。同样,TAM-R 细胞中的抗氧化剂水平也更高,从而提供了对辐照诱导的 ROS 的保护。抗氧化反应的主要激活剂 NRF2 在 TAM-R 细胞和接受短期术前他莫昔芬治疗的患者的肿瘤组织中有所增加。抑制NRF2可使TAM-R细胞对辐照重新敏感:结论:他莫昔芬诱导放射抗性的机制与细胞对持续增加的 ROS 水平的适应有关,这导致细胞的抗氧化能力和糖酵解能力长期处于上调状态。药物抑制抗氧化反应可使乳腺癌细胞对辐照重新敏感。
{"title":"Tamoxifen induces radioresistance through NRF2-mediated metabolic reprogramming in breast cancer.","authors":"F V Reinema, F C G J Sweep, G J Adema, W J M Peeters, J W M Martens, J Bussink, P N Span","doi":"10.1186/s40170-023-00304-4","DOIUrl":"10.1186/s40170-023-00304-4","url":null,"abstract":"<p><strong>Background: </strong>Recently, we reported that tamoxifen-resistant (TAM-R) breast cancer cells are cross-resistant to irradiation. Here, we investigated the mechanisms associated with tamoxifen-induced radioresistance, aiming to prevent or reverse resistance and improve breast cancer treatment.</p><p><strong>Methods: </strong>Wild-type ERα-positive MCF7 and ERα-negative MDA-MB-231 breast cancer cells and their TAM-R counterparts were analyzed for cellular metabolism using the Seahorse metabolic analyzer. Real-time ROS production, toxicity, and antioxidant capacity in response to H<sub>2</sub>O<sub>2</sub>, tamoxifen, and irradiation were determined. Tumor material from 28 breast cancer patients before and after short-term presurgical tamoxifen (ClinicalTrials.gov Identifier: NCT00738777, August 19, 2008) and cellular material was analyzed for NRF2 gene expression and immunohistochemistry. Re-sensitization of TAM-R cells to irradiation was established using pharmacological inhibition.</p><p><strong>Results: </strong>TAM-R cells exhibited decreased oxygen consumption and increased glycolysis, suggesting mitochondrial dysfunction. However, this did not explain radioresistance, as cells without mitochondria (Rho-0) were actually more radiosensitive. Real-time measurement of ROS after tamoxifen and H<sub>2</sub>O<sub>2</sub> exposure indicated lower ROS levels and toxicity in TAM-R cells. Consistently, higher antioxidant levels were found in TAM-R cells, providing protection from irradiation-induced ROS. NRF2, a main activator of the antioxidant response, was increased in TAM-R cells and in tumor tissue of patients treated with short-term presurgical tamoxifen. NRF2 inhibition re-sensitized TAM-R cells to irradiation.</p><p><strong>Conclusion: </strong>Mechanisms underlying tamoxifen-induced radioresistance are linked to cellular adaptations to persistently increased ROS levels, leading to cells with chronically upregulated antioxidant capacity and glycolysis. Pharmacological inhibition of antioxidant responses re-sensitizes breast cancer cells to irradiation.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"11 1","pages":"3"},"PeriodicalIF":6.0,"publicationDate":"2023-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9909892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10684458","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic profiling reveals metabolic features of consolidation therapy in pediatric acute lymphoblastic leukemia. 代谢谱揭示了儿童急性淋巴细胞白血病巩固治疗的代谢特征。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2023-01-23 DOI: 10.1186/s40170-023-00302-6
Jinqiu Fu, Aijun Zhang, Qinqin Liu, Dong Li, Xiaoming Wang, Libo Si

Acute lymphoblastic leukemia (ALL) and its treatment continue to pose substantial risks. To understand ALL more deeply, the metabolome in fasting plasma of 27 ALL patients before and after high-dose methotrexate therapies (consolidation therapy) including methotrexate and 6-mercaptopurine (6-MP) was investigated. Plasma metabolites were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS). Orthogonal projections to latent structures discriminant analysis and significance analysis of microarrays were used to evaluate the metabolic changes. Pathway enrichment and co-expression network analyses were performed to identify clusters of molecules, and 2826 metabolites were identified. Among them, 38 metabolites were identified by univariate analysis, and 7 metabolites that were altered by conditioning therapy were identified by multivariate analysis. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used for pathway enrichment analysis. Among the enriched KEGG pathways, the 3 significantly altered metabolic pathways were pyrimidine metabolism; phenylalanine, tyrosine, and tryptophan biosynthesis; and phenylalanine metabolism. In addition, L-phenylalanine was significantly correlated with blood urea nitrogen (BUN), and palmitoylcarnitine was correlated with aspartate aminotransferase (AST). In summary, consolidation therapy significantly affected pyrimidine- and phenylalanine-associated metabolic pathways in pediatric ALL patients. These findings may provide an insight into the role of metabolic profiling in consolidation treatment and as a potential for pediatric ALL patients.

急性淋巴细胞白血病(ALL)及其治疗仍然存在重大风险。为了更深入地了解ALL,我们研究了27例ALL患者在甲氨蝶呤和6-巯基嘌呤(6-MP)等高剂量甲氨蝶呤治疗(巩固治疗)前后空腹血浆代谢组的变化。采用液相色谱-串联质谱(LC-MS)分析血浆代谢物。利用微阵列的潜在结构正交投影判别分析和显著性分析来评估代谢变化。通过途径富集和共表达网络分析来鉴定分子簇,鉴定出2826种代谢物。其中单因素分析鉴定出38种代谢物,多因素分析鉴定出7种因调理治疗而改变的代谢物。利用京都基因与基因组百科全书(KEGG)数据库进行途径富集分析。在富集的KEGG途径中,3条代谢途径发生显著变化的是嘧啶代谢途径;苯丙氨酸、酪氨酸和色氨酸的生物合成;还有苯丙氨酸代谢。l -苯丙氨酸与血尿素氮(BUN)显著相关,棕榈酰肉碱与天冬氨酸转氨酶(AST)显著相关。总之,巩固治疗显著影响了小儿ALL患者嘧啶和苯丙氨酸相关的代谢途径。这些发现可能有助于了解代谢谱分析在巩固治疗中的作用,并为儿科ALL患者提供潜在的治疗方法。
{"title":"Metabolic profiling reveals metabolic features of consolidation therapy in pediatric acute lymphoblastic leukemia.","authors":"Jinqiu Fu,&nbsp;Aijun Zhang,&nbsp;Qinqin Liu,&nbsp;Dong Li,&nbsp;Xiaoming Wang,&nbsp;Libo Si","doi":"10.1186/s40170-023-00302-6","DOIUrl":"https://doi.org/10.1186/s40170-023-00302-6","url":null,"abstract":"<p><p>Acute lymphoblastic leukemia (ALL) and its treatment continue to pose substantial risks. To understand ALL more deeply, the metabolome in fasting plasma of 27 ALL patients before and after high-dose methotrexate therapies (consolidation therapy) including methotrexate and 6-mercaptopurine (6-MP) was investigated. Plasma metabolites were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS). Orthogonal projections to latent structures discriminant analysis and significance analysis of microarrays were used to evaluate the metabolic changes. Pathway enrichment and co-expression network analyses were performed to identify clusters of molecules, and 2826 metabolites were identified. Among them, 38 metabolites were identified by univariate analysis, and 7 metabolites that were altered by conditioning therapy were identified by multivariate analysis. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used for pathway enrichment analysis. Among the enriched KEGG pathways, the 3 significantly altered metabolic pathways were pyrimidine metabolism; phenylalanine, tyrosine, and tryptophan biosynthesis; and phenylalanine metabolism. In addition, L-phenylalanine was significantly correlated with blood urea nitrogen (BUN), and palmitoylcarnitine was correlated with aspartate aminotransferase (AST). In summary, consolidation therapy significantly affected pyrimidine- and phenylalanine-associated metabolic pathways in pediatric ALL patients. These findings may provide an insight into the role of metabolic profiling in consolidation treatment and as a potential for pediatric ALL patients.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"11 1","pages":"2"},"PeriodicalIF":5.9,"publicationDate":"2023-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9869545/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10667406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Arginase-1 inhibition reduces migration ability and metastatic colonization of colon cancer cells. 精氨酸酶-1抑制降低结肠癌细胞的迁移能力和转移定植。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2023-01-13 DOI: 10.1186/s40170-022-00301-z
Xiangdong Wang, Huihui Xiang, Yujiro Toyoshima, Weidong Shen, Shunsuke Shichi, Hiroki Nakamoto, Saori Kimura, Ko Sugiyama, Shigenori Homma, Yohei Miyagi, Akinobu Taketomi, Hidemitsu Kitamura

Background: Arginase-1 (ARG1), a urea cycle-related enzyme, catalyzes the hydrolysis of arginine to urea and ornithine, which regulates the proliferation, differentiation, and function of various cells. However, it is unclear whether ARG1 controls the progression and malignant alterations of colon cancer.

Methods: We established metastatic colonization mouse model and ARG1 overexpressing murine colon cancer CT26 cells to investigate whether activation of ARG1 was related to malignancy of colon cancer cells in vivo. Living cell numbers and migration ability of CT26 cells were evaluated in the presence of ARG inhibitor in vitro.

Results: Inhibition of arginase activity significantly suppressed the proliferation and migration ability of CT26 murine colon cancer cells in vitro. Overexpression of ARG1 in CT26 cells reduced intracellular L-arginine levels, enhanced cell migration, and promoted epithelial-mesenchymal transition. Metastatic colonization of CT26 cells in lung and liver tissues was significantly augmented by ARG1 overexpression in vivo. ARG1 gene expression was higher in the tumor tissues of liver metastasis than those of primary tumor, and arginase inhibition suppressed the migration ability of HCT116 human colon cancer cells.

Conclusion: Activation of ARG1 is related to the migration ability and metastatic colonization of colon cancer cells, and blockade of this process may be a novel strategy for controlling cancer malignancy.

背景:精氨酸酶-1 (Arginase-1, ARG1)是一种尿素循环相关酶,可催化精氨酸水解为尿素和鸟氨酸,调节多种细胞的增殖、分化和功能。然而,ARG1是否控制结肠癌的进展和恶性改变尚不清楚。方法:建立转移性定植小鼠模型和过表达ARG1的小鼠结肠癌CT26细胞,在体内研究ARG1的激活是否与结肠癌细胞的恶性化有关。体外观察ARG抑制剂作用下CT26细胞的活细胞数量和迁移能力。结果:抑制精氨酸酶活性可显著抑制CT26小鼠结肠癌细胞的体外增殖和迁移能力。ARG1在CT26细胞中的过表达降低了细胞内l-精氨酸水平,增强了细胞迁移,促进了上皮-间质转化。体内ARG1过表达可显著增强CT26细胞在肺和肝组织中的转移定植。ARG1基因在肝转移肿瘤组织中的表达高于原发肿瘤组织,精氨酸酶的抑制抑制了HCT116人结肠癌细胞的迁移能力。结论:ARG1的激活与结肠癌细胞的迁移能力和转移定殖有关,阻断这一过程可能是控制肿瘤恶性的新策略。
{"title":"Arginase-1 inhibition reduces migration ability and metastatic colonization of colon cancer cells.","authors":"Xiangdong Wang,&nbsp;Huihui Xiang,&nbsp;Yujiro Toyoshima,&nbsp;Weidong Shen,&nbsp;Shunsuke Shichi,&nbsp;Hiroki Nakamoto,&nbsp;Saori Kimura,&nbsp;Ko Sugiyama,&nbsp;Shigenori Homma,&nbsp;Yohei Miyagi,&nbsp;Akinobu Taketomi,&nbsp;Hidemitsu Kitamura","doi":"10.1186/s40170-022-00301-z","DOIUrl":"https://doi.org/10.1186/s40170-022-00301-z","url":null,"abstract":"<p><strong>Background: </strong>Arginase-1 (ARG1), a urea cycle-related enzyme, catalyzes the hydrolysis of arginine to urea and ornithine, which regulates the proliferation, differentiation, and function of various cells. However, it is unclear whether ARG1 controls the progression and malignant alterations of colon cancer.</p><p><strong>Methods: </strong>We established metastatic colonization mouse model and ARG1 overexpressing murine colon cancer CT26 cells to investigate whether activation of ARG1 was related to malignancy of colon cancer cells in vivo. Living cell numbers and migration ability of CT26 cells were evaluated in the presence of ARG inhibitor in vitro.</p><p><strong>Results: </strong>Inhibition of arginase activity significantly suppressed the proliferation and migration ability of CT26 murine colon cancer cells in vitro. Overexpression of ARG1 in CT26 cells reduced intracellular L-arginine levels, enhanced cell migration, and promoted epithelial-mesenchymal transition. Metastatic colonization of CT26 cells in lung and liver tissues was significantly augmented by ARG1 overexpression in vivo. ARG1 gene expression was higher in the tumor tissues of liver metastasis than those of primary tumor, and arginase inhibition suppressed the migration ability of HCT116 human colon cancer cells.</p><p><strong>Conclusion: </strong>Activation of ARG1 is related to the migration ability and metastatic colonization of colon cancer cells, and blockade of this process may be a novel strategy for controlling cancer malignancy.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"11 1","pages":"1"},"PeriodicalIF":5.9,"publicationDate":"2023-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9838026/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9080180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Functional noninvasive detection of glycolytic pancreatic ductal adenocarcinoma. 糖酵解性胰腺导管腺癌的功能无创检测。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-12-09 DOI: 10.1186/s40170-022-00298-5
Irina Heid, Corinna Münch, Sinan Karakaya, Smiths S Lueong, Alina M Winkelkotte, Sven T Liffers, Laura Godfrey, Phyllis F Y Cheung, Konstantinos Savvatakis, Geoffrey J Topping, Florian Englert, Lukas Kritzner, Martin Grashei, Andrea Tannapfel, Richard Viebahn, Heiner Wolters, Waldemar Uhl, Deepak Vangala, Esther M M Smeets, Erik H J G Aarntzen, Daniel Rauh, Wilko Weichert, Jörg D Hoheisel, Stephan A Hahn, Franz Schilling, Rickmer Braren, Marija Trajkovic-Arsic, Jens T Siveke

Background: Pancreatic ductal adenocarcinoma (PDAC) lacks effective treatment options beyond chemotherapy. Although molecular subtypes such as classical and QM (quasi-mesenchymal)/basal-like with transcriptome-based distinct signatures have been identified, deduced therapeutic strategies and targets remain elusive. Gene expression data show enrichment of glycolytic genes in the more aggressive and therapy-resistant QM subtype. However, whether the glycolytic transcripts are translated into functional glycolysis that could further be explored for metabolic targeting in QM subtype is still not known.

Methods: We used different patient-derived PDAC model systems (conventional and primary patient-derived cells, patient-derived xenografts (PDX), and patient samples) and performed transcriptional and functional metabolic analysis. These included RNAseq and Illumina HT12 bead array, in vitro Seahorse metabolic flux assays and metabolic drug targeting, and in vivo hyperpolarized [1-13C]pyruvate and [1-13C]lactate magnetic resonance spectroscopy (HP-MRS) in PDAC xenografts.

Results: We found that glycolytic metabolic dependencies are not unambiguously functionally exposed in all QM PDACs. Metabolic analysis demonstrated functional metabolic heterogeneity in patient-derived primary cells and less so in conventional cell lines independent of molecular subtype. Importantly, we observed that the glycolytic product lactate is actively imported into the PDAC cells and used in mitochondrial oxidation in both classical and QM PDAC cells, although more actively in the QM cell lines. By using HP-MRS, we were able to noninvasively identify highly glycolytic PDAC xenografts by detecting the last glycolytic enzymatic step and prominent intra-tumoral [1-13C]pyruvate and [1-13C]lactate interconversion in vivo.

Conclusion: Our study adds functional metabolic phenotyping to transcriptome-based analysis and proposes a functional approach to identify highly glycolytic PDACs as candidates for antimetabolic therapeutic avenues.

背景:胰腺导管腺癌(PDAC)缺乏化疗以外的有效治疗选择。虽然分子亚型如经典和QM(准间充质)/基底样与转录组为基础的不同特征已经确定,推断的治疗策略和靶点仍然难以捉摸。基因表达数据显示,在更具侵袭性和治疗抗性的QM亚型中,糖酵解基因丰富。然而,糖酵解转录本是否被翻译成功能性糖酵解,从而进一步探索QM亚型的代谢靶向性,目前尚不清楚。方法:我们使用不同的患者来源的PDAC模型系统(常规和原代患者来源的细胞,患者来源的异种移植物(PDX)和患者样本)并进行转录和功能代谢分析。其中包括RNAseq和Illumina HT12头阵列,体外海马代谢通量测定和代谢药物靶向,以及PDAC异种移植物体内超极化[1-13C]丙酮酸和[1-13C]乳酸磁共振波谱(HP-MRS)。结果:我们发现糖酵解代谢依赖性在所有QM pdac中并不是明确的功能暴露。代谢分析表明,患者来源的原代细胞的功能代谢异质性,而在独立于分子亚型的传统细胞系中则较少。重要的是,我们观察到糖酵解产物乳酸被积极地输入到PDAC细胞中,并在经典和QM PDAC细胞中用于线粒体氧化,尽管在QM细胞系中更为活跃。通过使用HP-MRS,我们能够通过检测最后一个糖酵解酶步骤和体内肿瘤内显著的[1-13C]丙酮酸和[1-13C]乳酸相互转化,无创地鉴定高度糖酵解的PDAC异种移植物。结论:我们的研究将功能代谢表型分析添加到基于转录组的分析中,并提出了一种功能方法来识别高度糖酵解的pdac作为抗代谢治疗途径的候选者。
{"title":"Functional noninvasive detection of glycolytic pancreatic ductal adenocarcinoma.","authors":"Irina Heid,&nbsp;Corinna Münch,&nbsp;Sinan Karakaya,&nbsp;Smiths S Lueong,&nbsp;Alina M Winkelkotte,&nbsp;Sven T Liffers,&nbsp;Laura Godfrey,&nbsp;Phyllis F Y Cheung,&nbsp;Konstantinos Savvatakis,&nbsp;Geoffrey J Topping,&nbsp;Florian Englert,&nbsp;Lukas Kritzner,&nbsp;Martin Grashei,&nbsp;Andrea Tannapfel,&nbsp;Richard Viebahn,&nbsp;Heiner Wolters,&nbsp;Waldemar Uhl,&nbsp;Deepak Vangala,&nbsp;Esther M M Smeets,&nbsp;Erik H J G Aarntzen,&nbsp;Daniel Rauh,&nbsp;Wilko Weichert,&nbsp;Jörg D Hoheisel,&nbsp;Stephan A Hahn,&nbsp;Franz Schilling,&nbsp;Rickmer Braren,&nbsp;Marija Trajkovic-Arsic,&nbsp;Jens T Siveke","doi":"10.1186/s40170-022-00298-5","DOIUrl":"https://doi.org/10.1186/s40170-022-00298-5","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) lacks effective treatment options beyond chemotherapy. Although molecular subtypes such as classical and QM (quasi-mesenchymal)/basal-like with transcriptome-based distinct signatures have been identified, deduced therapeutic strategies and targets remain elusive. Gene expression data show enrichment of glycolytic genes in the more aggressive and therapy-resistant QM subtype. However, whether the glycolytic transcripts are translated into functional glycolysis that could further be explored for metabolic targeting in QM subtype is still not known.</p><p><strong>Methods: </strong>We used different patient-derived PDAC model systems (conventional and primary patient-derived cells, patient-derived xenografts (PDX), and patient samples) and performed transcriptional and functional metabolic analysis. These included RNAseq and Illumina HT12 bead array, in vitro Seahorse metabolic flux assays and metabolic drug targeting, and in vivo hyperpolarized [1-<sup>13</sup>C]pyruvate and [1-<sup>13</sup>C]lactate magnetic resonance spectroscopy (HP-MRS) in PDAC xenografts.</p><p><strong>Results: </strong>We found that glycolytic metabolic dependencies are not unambiguously functionally exposed in all QM PDACs. Metabolic analysis demonstrated functional metabolic heterogeneity in patient-derived primary cells and less so in conventional cell lines independent of molecular subtype. Importantly, we observed that the glycolytic product lactate is actively imported into the PDAC cells and used in mitochondrial oxidation in both classical and QM PDAC cells, although more actively in the QM cell lines. By using HP-MRS, we were able to noninvasively identify highly glycolytic PDAC xenografts by detecting the last glycolytic enzymatic step and prominent intra-tumoral [1-<sup>13</sup>C]pyruvate and [1-<sup>13</sup>C]lactate interconversion in vivo.</p><p><strong>Conclusion: </strong>Our study adds functional metabolic phenotyping to transcriptome-based analysis and proposes a functional approach to identify highly glycolytic PDACs as candidates for antimetabolic therapeutic avenues.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"24"},"PeriodicalIF":5.9,"publicationDate":"2022-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9737747/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10329732","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PDK1- and PDK2-mediated metabolic reprogramming contributes to the TGFβ1-promoted stem-like properties in head and neck cancer. PDK1-和pdk2介导的代谢重编程有助于tgf β1促进头颈癌的茎样特性。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-12-06 DOI: 10.1186/s40170-022-00300-0
Wan-Hsuan Sun, Yun-Hsuan Chen, Hou-Hsuan Lee, Yu-Wen Tang, Kuang-Hui Sun

Background: Resistance to chemotherapeutic drugs is a key factor for cancer recurrence and metastases in head and neck cancer (HNC). Cancer stem cells (CSCs) in tumors have self-renewal, differentiation, and higher drug resistance capabilities, resulting in a poor prognosis for patients. In glucose metabolism, pyruvate dehydrogenase kinase (PDK) inhibits pyruvate dehydrogenase and impedes pyruvate from being metabolized into acetyl-CoA and entering the tricarboxylic acid cycle to generate energy. Studies have reported that PDK1 and PDK2 inhibition suppresses the growth, motility, and drug resistance of cancer cells. Furthermore, while TGFβ1 levels are persistently elevated in HNC patients with poor prognosis, the role of PDK isoforms in the TGFβ1-promoted progression and stem-like properties of HNC is unclear.

Methods: Levels of PDK1 and PDK2 were evaluated in HNC tissue microarrays by immunohistochemistry to explore potential clinical relevance. PDK1 and PDK2 were knocked down by the lentivirus shRNA system to investigate their role in TGFβ1-promoted tumor progression in vitro.

Results: We found that PDK2 levels were increased in the later stage of HNC tissues compared to constant PDK1 expression. After PDK1 and PDK2 knockdown, we discovered increased ATP production and decreased lactate production in TGFβ1-treated and untreated HNC cells. However, only PDK2 silencing significantly inhibited the clonogenic ability of HNC cells. We subsequently found that TGFβ1-promoted migration and invasion capabilities were decreased in PDK1 and PDK2 knockdown cells. The tumor spheroid-forming capability, motility, CSC genes, and multidrug-resistant genes were downregulated in PDK1 and PDK2 silencing CSCs. PDK1 and PDK2 inhibition reversed cisplatin and gemcitabine resistance of CSCs, but not paclitaxel resistance.

Conclusion: The results demonstrated that the PDK1- and PDK2-mediated Warburg effect contributes to the TGFβ1-enhanced stemness properties of HNC. Therefore, PDK1 and PDK2 may serve as molecular targets for the combination therapy of HNC.

背景:化疗药物耐药是头颈癌(HNC)复发和转移的关键因素。肿瘤中的肿瘤干细胞(Cancer stem cells, CSCs)具有自我更新、分化和较高的耐药能力,导致患者预后较差。在葡萄糖代谢中,丙酮酸脱氢酶激酶(PDK)抑制丙酮酸脱氢酶,阻碍丙酮酸代谢成乙酰辅酶a进入三羧酸循环产生能量。有研究报道,抑制PDK1和PDK2可抑制癌细胞的生长、运动和耐药性。此外,尽管tgf - β1水平在预后不良的HNC患者中持续升高,但PDK亚型在tgf - β1促进HNC进展和干细胞样特性中的作用尚不清楚。方法:采用免疫组织化学方法评估HNC组织微阵列中PDK1和PDK2的水平,以探讨其潜在的临床意义。通过慢病毒shRNA系统敲除PDK1和PDK2,研究它们在体外tgf β1促进肿瘤进展中的作用。结果:我们发现,与PDK1表达不变相比,在HNC组织的晚期,PDK2水平升高。在敲除PDK1和PDK2后,我们发现tgf - β1处理和未处理的HNC细胞中ATP的产生增加,乳酸的产生减少。然而,只有PDK2沉默显著抑制HNC细胞的克隆生成能力。我们随后发现tgf β1促进的迁移和侵袭能力在PDK1和PDK2敲低的细胞中下降。在PDK1和PDK2沉默的CSC中,肿瘤球体形成能力、运动性、CSC基因和多药耐药基因下调。PDK1和PDK2抑制逆转了CSCs对顺铂和吉西他滨的耐药,但没有逆转紫杉醇耐药。结论:PDK1-和pdk2介导的Warburg效应参与了tgf - β1增强HNC的干性特性。因此,PDK1和PDK2可能作为HNC联合治疗的分子靶点。
{"title":"PDK1- and PDK2-mediated metabolic reprogramming contributes to the TGFβ1-promoted stem-like properties in head and neck cancer.","authors":"Wan-Hsuan Sun,&nbsp;Yun-Hsuan Chen,&nbsp;Hou-Hsuan Lee,&nbsp;Yu-Wen Tang,&nbsp;Kuang-Hui Sun","doi":"10.1186/s40170-022-00300-0","DOIUrl":"https://doi.org/10.1186/s40170-022-00300-0","url":null,"abstract":"<p><strong>Background: </strong>Resistance to chemotherapeutic drugs is a key factor for cancer recurrence and metastases in head and neck cancer (HNC). Cancer stem cells (CSCs) in tumors have self-renewal, differentiation, and higher drug resistance capabilities, resulting in a poor prognosis for patients. In glucose metabolism, pyruvate dehydrogenase kinase (PDK) inhibits pyruvate dehydrogenase and impedes pyruvate from being metabolized into acetyl-CoA and entering the tricarboxylic acid cycle to generate energy. Studies have reported that PDK1 and PDK2 inhibition suppresses the growth, motility, and drug resistance of cancer cells. Furthermore, while TGFβ1 levels are persistently elevated in HNC patients with poor prognosis, the role of PDK isoforms in the TGFβ1-promoted progression and stem-like properties of HNC is unclear.</p><p><strong>Methods: </strong>Levels of PDK1 and PDK2 were evaluated in HNC tissue microarrays by immunohistochemistry to explore potential clinical relevance. PDK1 and PDK2 were knocked down by the lentivirus shRNA system to investigate their role in TGFβ1-promoted tumor progression in vitro.</p><p><strong>Results: </strong>We found that PDK2 levels were increased in the later stage of HNC tissues compared to constant PDK1 expression. After PDK1 and PDK2 knockdown, we discovered increased ATP production and decreased lactate production in TGFβ1-treated and untreated HNC cells. However, only PDK2 silencing significantly inhibited the clonogenic ability of HNC cells. We subsequently found that TGFβ1-promoted migration and invasion capabilities were decreased in PDK1 and PDK2 knockdown cells. The tumor spheroid-forming capability, motility, CSC genes, and multidrug-resistant genes were downregulated in PDK1 and PDK2 silencing CSCs. PDK1 and PDK2 inhibition reversed cisplatin and gemcitabine resistance of CSCs, but not paclitaxel resistance.</p><p><strong>Conclusion: </strong>The results demonstrated that the PDK1- and PDK2-mediated Warburg effect contributes to the TGFβ1-enhanced stemness properties of HNC. Therefore, PDK1 and PDK2 may serve as molecular targets for the combination therapy of HNC.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"23"},"PeriodicalIF":5.9,"publicationDate":"2022-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9727917/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10430523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Tadalafil increases the antitumor activity of 5-FU through inhibiting PRMT5-mediated glycolysis and cell proliferation in colorectal cancer. 他达拉非通过抑制prmt5介导的糖酵解和结直肠癌细胞增殖来提高5-FU的抗肿瘤活性。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-12-06 DOI: 10.1186/s40170-022-00299-4
Yao Shen, Pan Zhao, Kewei Dong, Jiajia Wang, Huichen Li, Mengyang Li, Ruikai Li, Suning Chen, Yuxia Shen, Zhiyu Liu, Mianjiao Xie, Peng Shen, Jian Zhang

Background: Protein arginine methyltransferase 5 (PRMT5) is upregulated in multiple tumors and plays a pivotal role in cancer cell proliferation. However, the role of PRMT5 in colorectal cancer remains poorly understood.

Methods: We detected the expression level of PRMT5 and glycolytic enzymes using online databases and colorectal cancer cell lines by immunohistochemical staining, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting. And MTT and colony formation assays were conducted to investigate cell proliferation. Then, we evaluated ECAR and OCR levels using a biological energy analyzer to investigate the energy status of colorectal cancer, and the transcriptional regulation was detected by dual luciferase reporter assay and ChIP assay. Finally, the efficacy of combined treatment of tadalafil and 5-FU was verified.

Results: PRMT5 was highly expressed in colorectal cancer tissues compared with their normal counterparts and correlated with poor prognosis in CRC patients. Then, we demonstrated that PRMT5 knockdown or loss of function attenuated the viability of CRC cells, while overexpression of PRMT5 promoted cell proliferation. Mechanistically, PRMT5 enhanced glycolysis through transcriptionally activating LDHA expression. In addition, the PRMT5 inhibitor, tadalafil, rendered CRC cells sensitive to antitumor agent 5-FU in vitro and in vivo.

Conclusions: Our data indicates that PRMT5 promoted colorectal cancer proliferation partially through activating glycolysis and may be a potential target for colorectal cancer therapy.

背景:蛋白精氨酸甲基转移酶5 (Protein arginine methyltransferase 5, PRMT5)在多种肿瘤中表达上调,在癌细胞增殖中起关键作用。然而,PRMT5在结直肠癌中的作用仍然知之甚少。方法:利用在线数据库和结直肠癌细胞系,采用免疫组化染色、定量实时聚合酶链反应(qRT-PCR)和western blotting检测PRMT5和糖酵解酶的表达水平。用MTT和菌落形成法观察细胞增殖情况。然后,我们使用生物能量分析仪评估ECAR和OCR水平,研究结直肠癌的能量状态,并通过双荧光素酶报告基因试验和ChIP试验检测转录调控。最后验证他达拉非与5-FU联合治疗的疗效。结果:PRMT5在结直肠癌组织中较正常组织高表达,且与结直肠癌患者预后不良相关。然后,我们证明PRMT5敲低或功能丧失会降低CRC细胞的活力,而PRMT5的过表达会促进细胞增殖。在机制上,PRMT5通过转录激活LDHA表达来增强糖酵解。此外,PRMT5抑制剂他达拉非在体外和体内均使CRC细胞对抗肿瘤药物5-FU敏感。结论:我们的数据表明PRMT5通过激活糖酵解部分促进结直肠癌的增殖,可能是结直肠癌治疗的潜在靶点。
{"title":"Tadalafil increases the antitumor activity of 5-FU through inhibiting PRMT5-mediated glycolysis and cell proliferation in colorectal cancer.","authors":"Yao Shen,&nbsp;Pan Zhao,&nbsp;Kewei Dong,&nbsp;Jiajia Wang,&nbsp;Huichen Li,&nbsp;Mengyang Li,&nbsp;Ruikai Li,&nbsp;Suning Chen,&nbsp;Yuxia Shen,&nbsp;Zhiyu Liu,&nbsp;Mianjiao Xie,&nbsp;Peng Shen,&nbsp;Jian Zhang","doi":"10.1186/s40170-022-00299-4","DOIUrl":"https://doi.org/10.1186/s40170-022-00299-4","url":null,"abstract":"<p><strong>Background: </strong>Protein arginine methyltransferase 5 (PRMT5) is upregulated in multiple tumors and plays a pivotal role in cancer cell proliferation. However, the role of PRMT5 in colorectal cancer remains poorly understood.</p><p><strong>Methods: </strong>We detected the expression level of PRMT5 and glycolytic enzymes using online databases and colorectal cancer cell lines by immunohistochemical staining, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting. And MTT and colony formation assays were conducted to investigate cell proliferation. Then, we evaluated ECAR and OCR levels using a biological energy analyzer to investigate the energy status of colorectal cancer, and the transcriptional regulation was detected by dual luciferase reporter assay and ChIP assay. Finally, the efficacy of combined treatment of tadalafil and 5-FU was verified.</p><p><strong>Results: </strong>PRMT5 was highly expressed in colorectal cancer tissues compared with their normal counterparts and correlated with poor prognosis in CRC patients. Then, we demonstrated that PRMT5 knockdown or loss of function attenuated the viability of CRC cells, while overexpression of PRMT5 promoted cell proliferation. Mechanistically, PRMT5 enhanced glycolysis through transcriptionally activating LDHA expression. In addition, the PRMT5 inhibitor, tadalafil, rendered CRC cells sensitive to antitumor agent 5-FU in vitro and in vivo.</p><p><strong>Conclusions: </strong>Our data indicates that PRMT5 promoted colorectal cancer proliferation partially through activating glycolysis and may be a potential target for colorectal cancer therapy.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"22"},"PeriodicalIF":5.9,"publicationDate":"2022-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9727889/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10719378","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Isotope tracing reveals distinct substrate preference in murine melanoma subtypes with differing anti-tumor immunity. 同位素示踪揭示了具有不同抗肿瘤免疫的小鼠黑色素瘤亚型的不同底物偏好。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-12-01 DOI: 10.1186/s40170-022-00296-7
Xinyi Zhang, Alexandra A Halberstam, Wanling Zhu, Brooks P Leitner, Durga Thakral, Marcus W Bosenberg, Rachel J Perry

Background: Research about tumor "metabolic flexibility"-the ability of cells to toggle between preferred nutrients depending on the metabolic context-has largely focused on obesity-associated cancers. However, increasing evidence for a key role for nutrient competition in the tumor microenvironment, as well as for substrate regulation of immune function, suggests that substrate metabolism deserves reconsideration in immunogenic tumors that are not strongly associated with obesity.

Methods: We compare two murine models: immunologically cold YUMM1.7 and immunologically-hot YUMMER1.7. We utilize stable isotope and radioisotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics analyses to comprehensively probe substrate preference in YUMM1.7 and YUMMER1.7 cells, with a subset of studies on the impact of available metabolites across a panel of five additional melanoma cell lines. We analyze bulk RNA-seq data and identify increased expression of amino acid and glucose metabolism genes in YUMMER1.7. Finally, we analyze melanoma patient RNA-seq data to identify potential prognostic predictors rooted in metabolism.

Results: We demonstrate using stable isotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics that immunologically-hot melanoma utilizes more glutamine than immunologically-cold melanoma in vivo and in vitro. Analyses of human melanoma RNA-seq data demonstrate that glutamine transporter and other anaplerotic gene expression positively correlates with lymphocyte infiltration and function.

Conclusions: Here, we highlight the importance of understanding metabolism in non-obesity-associated cancers, such as melanoma. This work advances the understanding of the correlation between metabolism and immunogenicity in the tumor microenvironment and provides evidence supporting metabolic gene expression as potential prognostic factors of melanoma progression and may inform investigations of adjunctive metabolic therapy in melanoma.

Trial registration: Deidentified data from The Cancer Genome Atlas were analyzed.

背景:关于肿瘤“代谢灵活性”的研究——细胞根据代谢环境在首选营养素之间切换的能力——主要集中在肥胖相关的癌症上。然而,越来越多的证据表明,营养竞争在肿瘤微环境中的关键作用,以及底物对免疫功能的调节,表明底物代谢在与肥胖没有强烈关联的免疫原性肿瘤中值得重新考虑。方法:比较两种小鼠模型:免疫冷型YUMM1.7和免疫热型YUMMER1.7。我们利用基于稳定同位素和放射性同位素示踪剂的代谢通量研究以及基于气相色谱和液相色谱的代谢组学分析来全面探测YUMM1.7和YUMMER1.7细胞中的底物偏好,并对另外五种黑色素瘤细胞系的可用代谢物的影响进行了一部分研究。我们分析了大量RNA-seq数据,发现YUMMER1.7中氨基酸和葡萄糖代谢基因的表达增加。最后,我们分析了黑色素瘤患者的RNA-seq数据,以确定植根于代谢的潜在预后预测因子。结果:我们通过基于稳定同位素示踪剂的代谢通量研究以及基于气相色谱和液相色谱的代谢组学证明,在体内和体外,免疫热黑色素瘤比免疫冷黑色素瘤利用更多的谷氨酰胺。对人类黑色素瘤RNA-seq数据的分析表明,谷氨酰胺转运蛋白和其他突变基因的表达与淋巴细胞浸润和功能呈正相关。结论:在这里,我们强调了解代谢在非肥胖相关癌症(如黑色素瘤)中的重要性。这项工作促进了对肿瘤微环境中代谢与免疫原性之间相关性的理解,并提供了支持代谢基因表达作为黑色素瘤进展的潜在预后因素的证据,并可能为黑色素瘤辅助代谢治疗的研究提供信息。试验注册:分析来自癌症基因组图谱的未识别数据。
{"title":"Isotope tracing reveals distinct substrate preference in murine melanoma subtypes with differing anti-tumor immunity.","authors":"Xinyi Zhang,&nbsp;Alexandra A Halberstam,&nbsp;Wanling Zhu,&nbsp;Brooks P Leitner,&nbsp;Durga Thakral,&nbsp;Marcus W Bosenberg,&nbsp;Rachel J Perry","doi":"10.1186/s40170-022-00296-7","DOIUrl":"https://doi.org/10.1186/s40170-022-00296-7","url":null,"abstract":"<p><strong>Background: </strong>Research about tumor \"metabolic flexibility\"-the ability of cells to toggle between preferred nutrients depending on the metabolic context-has largely focused on obesity-associated cancers. However, increasing evidence for a key role for nutrient competition in the tumor microenvironment, as well as for substrate regulation of immune function, suggests that substrate metabolism deserves reconsideration in immunogenic tumors that are not strongly associated with obesity.</p><p><strong>Methods: </strong>We compare two murine models: immunologically cold YUMM1.7 and immunologically-hot YUMMER1.7. We utilize stable isotope and radioisotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics analyses to comprehensively probe substrate preference in YUMM1.7 and YUMMER1.7 cells, with a subset of studies on the impact of available metabolites across a panel of five additional melanoma cell lines. We analyze bulk RNA-seq data and identify increased expression of amino acid and glucose metabolism genes in YUMMER1.7. Finally, we analyze melanoma patient RNA-seq data to identify potential prognostic predictors rooted in metabolism.</p><p><strong>Results: </strong>We demonstrate using stable isotope tracer-based metabolic flux studies as well as gas and liquid chromatography-based metabolomics that immunologically-hot melanoma utilizes more glutamine than immunologically-cold melanoma in vivo and in vitro. Analyses of human melanoma RNA-seq data demonstrate that glutamine transporter and other anaplerotic gene expression positively correlates with lymphocyte infiltration and function.</p><p><strong>Conclusions: </strong>Here, we highlight the importance of understanding metabolism in non-obesity-associated cancers, such as melanoma. This work advances the understanding of the correlation between metabolism and immunogenicity in the tumor microenvironment and provides evidence supporting metabolic gene expression as potential prognostic factors of melanoma progression and may inform investigations of adjunctive metabolic therapy in melanoma.</p><p><strong>Trial registration: </strong>Deidentified data from The Cancer Genome Atlas were analyzed.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"21"},"PeriodicalIF":5.9,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9714036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10061099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Blocking lncRNA-SNHG16 sensitizes gastric cancer cells to 5-Fu through targeting the miR-506-3p-PTBP1-mediated glucose metabolism. 阻断lncRNA-SNHG16通过靶向mir -506-3p- ptbp1介导的葡萄糖代谢使胃癌细胞对5-Fu增敏。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-11-29 DOI: 10.1186/s40170-022-00293-w
Yan Ding, Sujie Gao, Jiabin Zheng, Xuebo Chen

Background: Gastric cancer (GC) is a commonly occurring human malignancy. The 5-fluorouracil (5-Fu) is a first-line anti-gastric cancer agent. However, a large number of GC patients developed 5-Fu resistance. Currently, the roles and molecular mechanisms of the lncRNA-SNHG16-modulated 5-Fu resistance in gastric cancer remain elusive.

Methods: Expressions of lncRNA, miRNA, and mRNA were detected by qRT-PCR and Western blot. RNA-RNA interaction was examined by RNA pull-down and luciferase assay. Cell viability and apoptosis rate under 5-Fu treatments were determined by MTT assay and Annexin V assay. The glycolysis rate of GC cells was evaluated by glucose uptake and ECAR.

Results: Here, we report that SNHG16 as well as PTBP1, which is an RNA-binding protein, are positively associated with 5-Fu resistance to gastric cancer. SNHG16 and PTBP1 were significantly upregulated in gastric tumors and cell lines. Silencing SNHG16 or PTBP1 effectively sensitized GC cells to 5-Fu. Furthermore, glucose metabolism was remarkedly elevated in 5-Fu-resistant GC cells. Under low glucose supply, 5-Fu-resistant cells displayed higher vulnerability than parental GC cells. Bioinformatic analysis and luciferase assay demonstrated that SNHG16 downregulated miR-506-3p by sponging it to form a ceRNA network. We identified PTBP1 as a direct target of miR-506-3p in GC cells. RNA-seq results unveiled that PTBP1 positively regulated expressions of multiple glycolysis enzymes, including GLUT1, HK2, and LDHA. Bioinformatic analysis illustrated the 3'UTRs of glycolysis enzymes contained multiple PTBP1 binding sites, which were further verified by RNA pull-down and RNA immunoprecipitation assays. Consequently, we demonstrated that PTBP1 upregulated the mRNAs of glycolysis enzymes via promoting their mRNA stabilities. Finally, in vivo xenograft experiments validated that blocking the SNHG16-mediated miR-506-3p-PTBP1 axis effectively limited 5-Fu-resistant GC cell originated-xenograft tumor growth under 5-Fu treatments.

Conclusions: Our study demonstrates molecular mechanisms of the SNHG16-mediated 5-Fu resistance of GC cells through modulating the miR-506-3p-PTBP1-glucose metabolism axis, presenting a promising approach for anti-chemoresistance therapy.

背景:胃癌是一种常见的人类恶性肿瘤。5-氟尿嘧啶(5-Fu)是一线抗胃癌药物。然而,大量GC患者出现5-Fu耐药。目前,lncrna - snhg16调控的5-Fu耐药在胃癌中的作用和分子机制尚不明确。方法:采用qRT-PCR和Western blot检测lncRNA、miRNA和mRNA的表达。采用RNA下拉法和荧光素酶法检测RNA-RNA相互作用。MTT法和Annexin V法检测5-Fu处理下细胞活力和凋亡率。葡萄糖摄取和ECAR测定GC细胞的糖酵解速率。结果:我们报道了SNHG16和rna结合蛋白PTBP1与5-Fu对胃癌的耐药呈正相关。SNHG16和PTBP1在胃肿瘤和细胞系中显著上调。沉默SNHG16或PTBP1可有效使GC细胞对5-Fu敏感。此外,葡萄糖代谢在5- fu耐药的GC细胞中再次显著升高。在低糖条件下,5- fu耐药细胞比亲本GC细胞表现出更高的脆弱性。生物信息学分析和荧光素酶测定表明,SNHG16通过海绵化miR-506-3p形成ceRNA网络,从而下调miR-506-3p。我们发现PTBP1是GC细胞中miR-506-3p的直接靶点。RNA-seq结果显示,PTBP1正调控多种糖酵解酶的表达,包括GLUT1、HK2和LDHA。生物信息学分析表明,糖酵解酶的3′utr含有多个PTBP1结合位点,并通过RNA pull-down和RNA免疫沉淀实验进一步验证。因此,我们证明了PTBP1通过促进糖酵解酶mRNA的稳定性来上调糖酵解酶的mRNA。最后,体内异种移植实验证实,阻断snhg16介导的miR-506-3p-PTBP1轴可有效限制5-Fu治疗下5-Fu耐药GC细胞源异种移植肿瘤的生长。结论:我们的研究通过调节mir -506-3p- ptbp1 -葡萄糖代谢轴,揭示了snhg16介导的GC细胞5-Fu耐药的分子机制,为抗化疗耐药提供了一条有前景的途径。
{"title":"Blocking lncRNA-SNHG16 sensitizes gastric cancer cells to 5-Fu through targeting the miR-506-3p-PTBP1-mediated glucose metabolism.","authors":"Yan Ding,&nbsp;Sujie Gao,&nbsp;Jiabin Zheng,&nbsp;Xuebo Chen","doi":"10.1186/s40170-022-00293-w","DOIUrl":"https://doi.org/10.1186/s40170-022-00293-w","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is a commonly occurring human malignancy. The 5-fluorouracil (5-Fu) is a first-line anti-gastric cancer agent. However, a large number of GC patients developed 5-Fu resistance. Currently, the roles and molecular mechanisms of the lncRNA-SNHG16-modulated 5-Fu resistance in gastric cancer remain elusive.</p><p><strong>Methods: </strong>Expressions of lncRNA, miRNA, and mRNA were detected by qRT-PCR and Western blot. RNA-RNA interaction was examined by RNA pull-down and luciferase assay. Cell viability and apoptosis rate under 5-Fu treatments were determined by MTT assay and Annexin V assay. The glycolysis rate of GC cells was evaluated by glucose uptake and ECAR.</p><p><strong>Results: </strong>Here, we report that SNHG16 as well as PTBP1, which is an RNA-binding protein, are positively associated with 5-Fu resistance to gastric cancer. SNHG16 and PTBP1 were significantly upregulated in gastric tumors and cell lines. Silencing SNHG16 or PTBP1 effectively sensitized GC cells to 5-Fu. Furthermore, glucose metabolism was remarkedly elevated in 5-Fu-resistant GC cells. Under low glucose supply, 5-Fu-resistant cells displayed higher vulnerability than parental GC cells. Bioinformatic analysis and luciferase assay demonstrated that SNHG16 downregulated miR-506-3p by sponging it to form a ceRNA network. We identified PTBP1 as a direct target of miR-506-3p in GC cells. RNA-seq results unveiled that PTBP1 positively regulated expressions of multiple glycolysis enzymes, including GLUT1, HK2, and LDHA. Bioinformatic analysis illustrated the 3'UTRs of glycolysis enzymes contained multiple PTBP1 binding sites, which were further verified by RNA pull-down and RNA immunoprecipitation assays. Consequently, we demonstrated that PTBP1 upregulated the mRNAs of glycolysis enzymes via promoting their mRNA stabilities. Finally, in vivo xenograft experiments validated that blocking the SNHG16-mediated miR-506-3p-PTBP1 axis effectively limited 5-Fu-resistant GC cell originated-xenograft tumor growth under 5-Fu treatments.</p><p><strong>Conclusions: </strong>Our study demonstrates molecular mechanisms of the SNHG16-mediated 5-Fu resistance of GC cells through modulating the miR-506-3p-PTBP1-glucose metabolism axis, presenting a promising approach for anti-chemoresistance therapy.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"20"},"PeriodicalIF":5.9,"publicationDate":"2022-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9707261/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10694564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Altered acetyl-CoA metabolism presents a new potential immunotherapy target in the obese lung microenvironment. 改变乙酰辅酶a代谢是肥胖肺微环境中一个新的潜在免疫治疗靶点。
IF 5.9 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-10-26 DOI: 10.1186/s40170-022-00292-x
Spencer R Rosario, Randall J Smith, Santosh K Patnaik, Song Liu, Joseph Barbi, Sai Yendamuri

Contrary to the "obesity paradox," which arises from retrospective studies relying on body mass index to define obesity, epidemiologic evidence suggests central or visceral obesity is associated with a higher risk for the development of lung cancer. About 60% of individuals at high risk for developing lung cancer or those already with early-stage disease are either overweight or obese. Findings from resected patient tumors and mouse lung tumor models show obesity dampens immune activity in the tumor microenvironment (TME) encouraging disease progression. In line with this, we have observed a marked, obesity-specific enhancement in the presence and phenotype of immunosuppressive regulatory T (Treg) cells in murine tumors as well as the airways of both humans and mice. Leveraging direct metabolomic measurements and robust inferred analyses from RNA-sequencing data, we here demonstrate for the first time that visceral adiposity alters the lung microenvironment via dysregulated acetyl-CoA metabolism in a direction that facilitates immune suppression and lung carcinogenesis.

与“肥胖悖论”相反,“肥胖悖论”源于依靠体重指数来定义肥胖的回顾性研究,流行病学证据表明,中枢性或内脏性肥胖与肺癌发展的高风险有关。大约60%的肺癌高危人群或已经患有早期疾病的人要么超重,要么肥胖。来自切除的患者肿瘤和小鼠肺肿瘤模型的研究结果显示,肥胖会抑制肿瘤微环境(TME)中的免疫活性,从而促进疾病进展。与此相一致,我们观察到小鼠肿瘤以及人类和小鼠气道中免疫抑制调节性T (Treg)细胞的存在和表型显着的肥胖特异性增强。利用直接代谢组学测量和来自rna测序数据的强大推断分析,我们在这里首次证明了内脏脂肪通过失调的乙酰辅酶a代谢改变肺微环境,从而促进免疫抑制和肺癌发生。
{"title":"Altered acetyl-CoA metabolism presents a new potential immunotherapy target in the obese lung microenvironment.","authors":"Spencer R Rosario,&nbsp;Randall J Smith,&nbsp;Santosh K Patnaik,&nbsp;Song Liu,&nbsp;Joseph Barbi,&nbsp;Sai Yendamuri","doi":"10.1186/s40170-022-00292-x","DOIUrl":"https://doi.org/10.1186/s40170-022-00292-x","url":null,"abstract":"<p><p>Contrary to the \"obesity paradox,\" which arises from retrospective studies relying on body mass index to define obesity, epidemiologic evidence suggests central or visceral obesity is associated with a higher risk for the development of lung cancer. About 60% of individuals at high risk for developing lung cancer or those already with early-stage disease are either overweight or obese. Findings from resected patient tumors and mouse lung tumor models show obesity dampens immune activity in the tumor microenvironment (TME) encouraging disease progression. In line with this, we have observed a marked, obesity-specific enhancement in the presence and phenotype of immunosuppressive regulatory T (Treg) cells in murine tumors as well as the airways of both humans and mice. Leveraging direct metabolomic measurements and robust inferred analyses from RNA-sequencing data, we here demonstrate for the first time that visceral adiposity alters the lung microenvironment via dysregulated acetyl-CoA metabolism in a direction that facilitates immune suppression and lung carcinogenesis.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"17"},"PeriodicalIF":5.9,"publicationDate":"2022-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9598035/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9620073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
Scaffold-mediated switching of lymphoma metabolism in culture. 支架介导的淋巴瘤培养新陈代谢转换。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2022-10-12 DOI: 10.1186/s40170-022-00291-y
Rachana Bhatt, Dashnamoorthy Ravi, Andrew M Evens, Biju Parekkadan

Background: Diffuse large B cell lymphoma (DLBCL) is an aggressive subtype of non-Hodgkin lymphoma (NHL) and accounts for about a third of all NHL cases. A significant proportion (~40%) of treated DLBCL patients develop refractory or relapsed disease due to drug resistance which can be attributed to metabolomic and genetic variations amongst diverse DLBCL subtypes. An assay platform that reproduces metabolic patterns of DLBCL in vivo could serve as a useful model for DLBCL.

Methods: This report investigated metabolic functions in 2D and 3D cell cultures using parental and drug-resistant DLBCL cell lines as compared to patient biopsy tissue.

Results: A 3D culture model controlled the proliferation of parental and drug-resistant DLBCL cell lines, SUDHL-10, SUDHL-10 RR (rituximab resistant), and SUDHL-10 OR (obinutuzumab resistant), as well as retained differential sensitivity to CHOP. The results from metabolic profiling and isotope tracer studies with D-glucose-13C6 indicated metabolic switching in 3D culture when compared with a 2D environment. Analysis of DLBCL patient tumor tissue revealed that the metabolic changes in 3D grown cells were shifted towards that of clinical specimens.

Conclusion: 3D culture restrained DLBCL cell line growth and modulated metabolic pathways that trend towards the biological characteristics of patient tumors. Counter-intuitively, this research thereby contends that 3D matrices can be a tool to control tumor function towards a slower growing and metabolically dormant state that better reflects in vivo tumor physiology.

背景:弥漫大B细胞淋巴瘤(DLBCL)是非霍奇金淋巴瘤(NHL)的一种侵袭性亚型,约占所有NHL病例的三分之一。在接受治疗的 DLBCL 患者中,有相当一部分(约 40%)因耐药性而出现难治或复发,这可能是由于不同 DLBCL 亚型之间的代谢组学和基因变异造成的。能再现 DLBCL 体内代谢模式的检测平台可作为 DLBCL 的有用模型:本报告使用亲代和耐药DLBCL细胞系与患者活检组织对比,研究了二维和三维细胞培养物的代谢功能:结果:三维培养模型控制了亲代和耐药 DLBCL 细胞系 SUDHL-10、SUDHL-10 RR(利妥昔单抗耐药)和 SUDHL-10 OR(奥比妥珠单抗耐药)的增殖,并保留了对 CHOP 的不同敏感性。使用 D-glucose-13C6 进行的代谢分析和同位素示踪研究结果表明,与二维环境相比,三维培养中的代谢发生了转换。结论:三维培养抑制了 DLBCL 细胞系的生长,并调节了代谢途径,使其趋向于患者肿瘤的生物学特征。结论:三维培养抑制了 DLBCL 细胞系的生长,并调节了代谢途径,使其趋向于患者肿瘤的生物学特征。与直觉相反,这项研究认为三维基质可以作为一种工具,控制肿瘤功能,使其趋向于生长缓慢、代谢休眠的状态,从而更好地反映体内肿瘤的生理机能。
{"title":"Scaffold-mediated switching of lymphoma metabolism in culture.","authors":"Rachana Bhatt, Dashnamoorthy Ravi, Andrew M Evens, Biju Parekkadan","doi":"10.1186/s40170-022-00291-y","DOIUrl":"10.1186/s40170-022-00291-y","url":null,"abstract":"<p><strong>Background: </strong>Diffuse large B cell lymphoma (DLBCL) is an aggressive subtype of non-Hodgkin lymphoma (NHL) and accounts for about a third of all NHL cases. A significant proportion (~40%) of treated DLBCL patients develop refractory or relapsed disease due to drug resistance which can be attributed to metabolomic and genetic variations amongst diverse DLBCL subtypes. An assay platform that reproduces metabolic patterns of DLBCL in vivo could serve as a useful model for DLBCL.</p><p><strong>Methods: </strong>This report investigated metabolic functions in 2D and 3D cell cultures using parental and drug-resistant DLBCL cell lines as compared to patient biopsy tissue.</p><p><strong>Results: </strong>A 3D culture model controlled the proliferation of parental and drug-resistant DLBCL cell lines, SUDHL-10, SUDHL-10 RR (rituximab resistant), and SUDHL-10 OR (obinutuzumab resistant), as well as retained differential sensitivity to CHOP. The results from metabolic profiling and isotope tracer studies with D-glucose-<sup>13</sup>C<sub>6</sub> indicated metabolic switching in 3D culture when compared with a 2D environment. Analysis of DLBCL patient tumor tissue revealed that the metabolic changes in 3D grown cells were shifted towards that of clinical specimens.</p><p><strong>Conclusion: </strong>3D culture restrained DLBCL cell line growth and modulated metabolic pathways that trend towards the biological characteristics of patient tumors. Counter-intuitively, this research thereby contends that 3D matrices can be a tool to control tumor function towards a slower growing and metabolically dormant state that better reflects in vivo tumor physiology.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"10 1","pages":"15"},"PeriodicalIF":6.0,"publicationDate":"2022-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9559005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9841008","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer & Metabolism
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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