首页 > 最新文献

Cancer & Metabolism最新文献

英文 中文
Hyperlipidemia drives tumor growth in a mouse model of obesity-accelerated breast cancer growth. 在肥胖小鼠模型中,高脂血症驱动肿瘤生长——加速乳腺癌生长。
IF 5.3 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-08-28 DOI: 10.1186/s40170-025-00407-0
Renan Fl Vieira, Sawyer R Sanchez, Menusha Arumugam, Peyton D Mower, Meghan C Curtin, Abigail E Jackson, Molly R Gallop, Jillian Wright, Alexis Bowles, Gregory S Ducker, Keren I Hilgendorf, Amandine Chaix

Obesity is an established risk factor for breast cancer (BC), yet the specific mechanisms driving this association remain unclear. Dysregulated lipid metabolism has emerged as a key factor in cancer cell biology, and, while obesity is often accompanied by hyperlipidemia, the isolated impact of elevated lipid levels on BC growth has not been experimentally tested. Using the E0771 and Py230 orthotopic models of obesity-accelerated BC growth in immune-competent mice, we investigated the role of systemic lipids on tumor growth. Combining dietary and genetic mouse models, we show that elevated circulating lipids are sufficient to accelerate BC tumor growth even in the absence of obesity or alterations in blood glucose and/or insulin levels. Pharmacological lowering of systemic lipid levels attenuates BC growth in obese mice, suggesting a direct role for lipids in fueling tumor expansion. Notably, we also show that weight loss alone, without a corresponding reduction in lipid levels such as that induced by a ketogenic diet, fails to protect against BC, highlighting the necessity of targeting lipid metabolism in obesity-associated BC. Our findings establish hyperlipidemia as a critical driver of BC progression and suggest that lipid-lowering interventions may be a promising strategy to mitigate BC risk in individuals with obesity.

肥胖是乳腺癌(BC)的一个确定的危险因素,但驱动这种关联的具体机制尚不清楚。脂质代谢失调已成为癌细胞生物学中的一个关键因素,虽然肥胖通常伴有高脂血症,但脂质水平升高对BC生长的单独影响尚未得到实验测试。利用E0771和Py230原位肥胖加速免疫能力小鼠BC生长模型,我们研究了全身脂质对肿瘤生长的作用。结合饮食和遗传小鼠模型,我们发现即使在没有肥胖或血糖和/或胰岛素水平改变的情况下,循环脂质升高也足以加速BC肿瘤的生长。药理学降低全身脂质水平可减缓肥胖小鼠的BC生长,提示脂质在促进肿瘤扩张中起直接作用。值得注意的是,我们还表明,单独减肥,没有相应的脂质水平降低,如由生酮饮食引起的,不能预防BC,强调了针对肥胖相关BC的脂质代谢的必要性。我们的研究结果确定高脂血症是BC进展的关键驱动因素,并表明降脂干预可能是减轻肥胖个体BC风险的一种有希望的策略。
{"title":"Hyperlipidemia drives tumor growth in a mouse model of obesity-accelerated breast cancer growth.","authors":"Renan Fl Vieira, Sawyer R Sanchez, Menusha Arumugam, Peyton D Mower, Meghan C Curtin, Abigail E Jackson, Molly R Gallop, Jillian Wright, Alexis Bowles, Gregory S Ducker, Keren I Hilgendorf, Amandine Chaix","doi":"10.1186/s40170-025-00407-0","DOIUrl":"10.1186/s40170-025-00407-0","url":null,"abstract":"<p><p>Obesity is an established risk factor for breast cancer (BC), yet the specific mechanisms driving this association remain unclear. Dysregulated lipid metabolism has emerged as a key factor in cancer cell biology, and, while obesity is often accompanied by hyperlipidemia, the isolated impact of elevated lipid levels on BC growth has not been experimentally tested. Using the E0771 and Py230 orthotopic models of obesity-accelerated BC growth in immune-competent mice, we investigated the role of systemic lipids on tumor growth. Combining dietary and genetic mouse models, we show that elevated circulating lipids are sufficient to accelerate BC tumor growth even in the absence of obesity or alterations in blood glucose and/or insulin levels. Pharmacological lowering of systemic lipid levels attenuates BC growth in obese mice, suggesting a direct role for lipids in fueling tumor expansion. Notably, we also show that weight loss alone, without a corresponding reduction in lipid levels such as that induced by a ketogenic diet, fails to protect against BC, highlighting the necessity of targeting lipid metabolism in obesity-associated BC. Our findings establish hyperlipidemia as a critical driver of BC progression and suggest that lipid-lowering interventions may be a promising strategy to mitigate BC risk in individuals with obesity.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"39"},"PeriodicalIF":5.3,"publicationDate":"2025-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12395885/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144943944","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
Glutaminase inhibition ameliorates cancer-associated fibroblast lipid support of pancreatic cancer cell growth. 谷氨酰胺酶抑制改善胰腺癌细胞生长的癌相关成纤维细胞脂质支持。
IF 5.3 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-08-20 DOI: 10.1186/s40170-025-00389-z
Xu Han, Laura C Kim, Nicholas P Lesner, Xuanyan Cai, Tran Ngoc Van Le, M Celeste Simon

Background: Lipid homeostasis is critical for pancreatic adenocarcinoma (PDAC) cell survival under hypoxic and nutrient-deprived conditions. Hypoxia inhibits unsaturated lipid biosynthesis, compelling cancer cells to depend on exogenous unsaturated lipids to counteract saturated lipid-induced toxicity. Our previous work revealed that cancer-associated fibroblasts (CAFs) secrete unsaturated lipids, primarily lysophosphatidylcholines (LPCs), to alleviate lipotoxic stress in PDAC cells. Here, we conducted a drug screen to identify compounds that bypass the rescue effect of exogenous LPCs on cancer cell survival under stress.

Methods: We employed high-throughput screening of a bioactive chemical library with 3,336 compounds, including FDA-approved drugs and drug-like molecules against defined molecular targets. Two assays were performed: a cytotoxicity assay to exclude indiscriminately toxic compounds at 1 μM and an LPC crosstalk inhibition assay to identify compounds that selectively reduce cancer cell viability in the presence of LPCs under stress conditions.

Results: CB-839, a glutaminase inhibitor, was identified as the most effective compound, selectively inhibiting the LPC-mediated rescue of PDAC cell viability effect without intrinsic cytotoxicity. Mechanistic studies revealed that CB-839 induces cell death by activating the pro-apoptotic ATF4/CHOP pathway, reducing antioxidant production, and increasing reactive oxygen species (ROS). While CB-839 showed limited efficacy against PDAC tumor cells alone in vivo, it modestly inhibited tumor growth in a PDAC-CAF co-implanted subcutaneous mouse model, highlighting its potential to disrupt CAF-mediated nutrient support. Additionally, glutamine antagonists showed more potent tumor-suppressive effects than CB-839.

Conclusion: Our findings emphasize the importance of glutamine metabolism inhibition in suppressing tumor growth and disrupting CAF-mediated crosstalk. We further underscore the potential of glutamine antagonist prodrugs as a strategy to target metabolic vulnerabilities in PDAC.

背景:在缺氧和营养缺乏的条件下,脂质稳态对胰腺腺癌(PDAC)细胞的存活至关重要。缺氧抑制不饱和脂质生物合成,迫使癌细胞依赖外源性不饱和脂质来抵消饱和脂质诱导的毒性。我们之前的研究表明,癌症相关成纤维细胞(CAFs)分泌不饱和脂质,主要是溶血磷脂酰胆碱(LPCs),以减轻PDAC细胞中的脂毒性应激。在这里,我们进行了药物筛选,以确定绕过外源性LPCs对应激下癌细胞存活的拯救作用的化合物。方法:采用高通量筛选方法,对3336种化合物进行生物活性化学文库筛选,包括fda批准的药物和针对特定分子靶点的药物样分子。研究人员进行了两项实验:一项是细胞毒性实验,以排除1 μM下不加区分的毒性化合物;另一项是LPC串扰抑制实验,以识别在应激条件下LPC存在时选择性降低癌细胞活力的化合物。结果:谷氨酰胺酶抑制剂CB-839被鉴定为最有效的化合物,可选择性抑制lpc介导的PDAC细胞活力恢复效应,而无内在的细胞毒性。机制研究表明,CB-839通过激活促凋亡的ATF4/CHOP通路,减少抗氧化剂的产生,增加活性氧(ROS)诱导细胞死亡。虽然CB-839在体内单独对PDAC肿瘤细胞的作用有限,但在PDAC- caf共植入皮下小鼠模型中,它适度抑制肿瘤生长,突出了其破坏cac介导的营养支持的潜力。此外,谷氨酰胺拮抗剂表现出比CB-839更有效的肿瘤抑制作用。结论:我们的研究结果强调了谷氨酰胺代谢抑制在抑制肿瘤生长和破坏cafa介导的串扰中的重要性。我们进一步强调谷氨酰胺拮抗剂前药作为针对PDAC代谢脆弱性的策略的潜力。
{"title":"Glutaminase inhibition ameliorates cancer-associated fibroblast lipid support of pancreatic cancer cell growth.","authors":"Xu Han, Laura C Kim, Nicholas P Lesner, Xuanyan Cai, Tran Ngoc Van Le, M Celeste Simon","doi":"10.1186/s40170-025-00389-z","DOIUrl":"10.1186/s40170-025-00389-z","url":null,"abstract":"<p><strong>Background: </strong>Lipid homeostasis is critical for pancreatic adenocarcinoma (PDAC) cell survival under hypoxic and nutrient-deprived conditions. Hypoxia inhibits unsaturated lipid biosynthesis, compelling cancer cells to depend on exogenous unsaturated lipids to counteract saturated lipid-induced toxicity. Our previous work revealed that cancer-associated fibroblasts (CAFs) secrete unsaturated lipids, primarily lysophosphatidylcholines (LPCs), to alleviate lipotoxic stress in PDAC cells. Here, we conducted a drug screen to identify compounds that bypass the rescue effect of exogenous LPCs on cancer cell survival under stress.</p><p><strong>Methods: </strong>We employed high-throughput screening of a bioactive chemical library with 3,336 compounds, including FDA-approved drugs and drug-like molecules against defined molecular targets. Two assays were performed: a cytotoxicity assay to exclude indiscriminately toxic compounds at 1 μM and an LPC crosstalk inhibition assay to identify compounds that selectively reduce cancer cell viability in the presence of LPCs under stress conditions.</p><p><strong>Results: </strong>CB-839, a glutaminase inhibitor, was identified as the most effective compound, selectively inhibiting the LPC-mediated rescue of PDAC cell viability effect without intrinsic cytotoxicity. Mechanistic studies revealed that CB-839 induces cell death by activating the pro-apoptotic ATF4/CHOP pathway, reducing antioxidant production, and increasing reactive oxygen species (ROS). While CB-839 showed limited efficacy against PDAC tumor cells alone in vivo, it modestly inhibited tumor growth in a PDAC-CAF co-implanted subcutaneous mouse model, highlighting its potential to disrupt CAF-mediated nutrient support. Additionally, glutamine antagonists showed more potent tumor-suppressive effects than CB-839.</p><p><strong>Conclusion: </strong>Our findings emphasize the importance of glutamine metabolism inhibition in suppressing tumor growth and disrupting CAF-mediated crosstalk. We further underscore the potential of glutamine antagonist prodrugs as a strategy to target metabolic vulnerabilities in PDAC.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"38"},"PeriodicalIF":5.3,"publicationDate":"2025-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366226/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144943962","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
NSUN2 promotes colorectal cancer progression by stabilizing PHGDH mRNA to promote serine metabolism reprogramming. NSUN2通过稳定PHGDH mRNA促进丝氨酸代谢重编程来促进结直肠癌的进展。
IF 5.3 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-08-14 DOI: 10.1186/s40170-025-00406-1
Hao Li, Tingyue Gong, Yongheng Zhao, Yang Luo, Shuibin Tang, Tingfeng Wang, Haiping Lin, Ming Zhong

Purpose: Cancer cells rely on serine biosynthesis for growth, but its regulation in colorectal cancer (CRC) remains not well understood. This study identifies the m5C methyltransferase NSUN2 (NOP2/Sun domain family, member 2) as a key regulator of serine biosynthesis, revealing a novel mechanism driving CRC progression.

Methods: The expression and prognostic value of NSUN2 were evaluated using bioinformatics analyses and immunohistochemistry (IHC) assays. The effects of NSUN2 on cellular serine biosynthesis, intracellular reactive oxygen species (ROS) levels, and apoptosis levels were analyzed both in vitro and in vivo. Additionally, RNA sequencing, Methylated RNA Immunoprecipitation sequencing (MeRIP-seq), RNA immunoprecipitation (RIP), and RNA stability assays were utilized to screen and validate the association between NSUN2 and phosphoglycerate dehydrogenase (PHGDH).

Results: NSUN2 was found to be highly expressed in CRC and associated with poor patient survival. PHGDH, a direct downstream target of NSUN2, plays a crucial role in NSUN2-mediated serine biosynthesis. Furthermore, inhibition of NSUN2 significantly reduced the intracellular NADH/NAD+ and NADPH/NADP+ ratios, leading to an increase in ROS levels and apoptosis levels, thereby inhibiting CRC progression. Additionally, NSUN2 enhances PHGDH expression and mRNA stability by binding to the "reader" protein m5C-Aly/REF export factor (ALYREF).

Conclusions: This study identified a novel NSUN2/ALYREF/m5C-PHGDH axis might be promising therapeutic targets for CRC.

目的:肿瘤细胞依赖丝氨酸生物合成来生长,但其在结直肠癌(CRC)中的调控尚不清楚。本研究发现m5C甲基转移酶NSUN2 (NOP2/Sun结构域家族,成员2)是丝氨酸生物合成的关键调节因子,揭示了驱动结直肠癌进展的新机制。方法:采用生物信息学分析和免疫组化(IHC)方法评价NSUN2的表达及预后价值。在体外和体内分析了NSUN2对细胞丝氨酸生物合成、细胞内活性氧(ROS)水平和凋亡水平的影响。此外,利用RNA测序、甲基化RNA免疫沉淀测序(MeRIP-seq)、RNA免疫沉淀(RIP)和RNA稳定性测定来筛选和验证NSUN2与磷酸甘油酸脱氢酶(PHGDH)之间的关联。结果:发现NSUN2在结直肠癌中高表达,并与患者生存差相关。PHGDH是NSUN2的直接下游靶点,在NSUN2介导的丝氨酸生物合成中起着至关重要的作用。此外,抑制NSUN2显著降低细胞内NADH/NAD+和NADPH/NADP+比值,导致ROS水平和凋亡水平升高,从而抑制结直肠癌的进展。此外,NSUN2通过结合“阅读器”蛋白m5C-Aly/REF输出因子(ALYREF)增强PHGDH表达和mRNA稳定性。结论:本研究确定了一个新的NSUN2/ALYREF/m5C-PHGDH轴可能是CRC的有希望的治疗靶点。
{"title":"NSUN2 promotes colorectal cancer progression by stabilizing PHGDH mRNA to promote serine metabolism reprogramming.","authors":"Hao Li, Tingyue Gong, Yongheng Zhao, Yang Luo, Shuibin Tang, Tingfeng Wang, Haiping Lin, Ming Zhong","doi":"10.1186/s40170-025-00406-1","DOIUrl":"10.1186/s40170-025-00406-1","url":null,"abstract":"<p><strong>Purpose: </strong>Cancer cells rely on serine biosynthesis for growth, but its regulation in colorectal cancer (CRC) remains not well understood. This study identifies the m<sup>5</sup>C methyltransferase NSUN2 (NOP2/Sun domain family, member 2) as a key regulator of serine biosynthesis, revealing a novel mechanism driving CRC progression.</p><p><strong>Methods: </strong>The expression and prognostic value of NSUN2 were evaluated using bioinformatics analyses and immunohistochemistry (IHC) assays. The effects of NSUN2 on cellular serine biosynthesis, intracellular reactive oxygen species (ROS) levels, and apoptosis levels were analyzed both in vitro and in vivo. Additionally, RNA sequencing, Methylated RNA Immunoprecipitation sequencing (MeRIP-seq), RNA immunoprecipitation (RIP), and RNA stability assays were utilized to screen and validate the association between NSUN2 and phosphoglycerate dehydrogenase (PHGDH).</p><p><strong>Results: </strong>NSUN2 was found to be highly expressed in CRC and associated with poor patient survival. PHGDH, a direct downstream target of NSUN2, plays a crucial role in NSUN2-mediated serine biosynthesis. Furthermore, inhibition of NSUN2 significantly reduced the intracellular NADH/NAD<sup>+</sup> and NADPH/NADP<sup>+</sup> ratios, leading to an increase in ROS levels and apoptosis levels, thereby inhibiting CRC progression. Additionally, NSUN2 enhances PHGDH expression and mRNA stability by binding to the \"reader\" protein m<sup>5</sup>C-Aly/REF export factor (ALYREF).</p><p><strong>Conclusions: </strong>This study identified a novel NSUN2/ALYREF/m<sup>5</sup>C-PHGDH axis might be promising therapeutic targets for CRC.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"37"},"PeriodicalIF":5.3,"publicationDate":"2025-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12351839/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144854655","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
Pharmacological inhibition of Peroxisome Proliferation-Activated Receptor Delta (PPARδ) imparts selective leukemia cell death. 过氧化物酶体增殖激活受体δ (PPARδ)的药物抑制可导致白血病细胞选择性死亡。
IF 5.3 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-07-25 DOI: 10.1186/s40170-025-00402-5
Yingying Yang, Ekaterina Parfenova, Nikolina Vrdoljak, Mark Minden, Jessica Luc, Andrew C Doxey, Paul A Spagnuolo

Background: Acute myeloid leukemia (AML) is a devastating hematological malignancy with limited therapeutic options and poor survival outcomes. Therefore, the development of novel and selective anti-AML therapies is needed. 6-methoxydihydroavicine (6ME), a benzophenanthridine alkaloid, imparted selective AML cell death in vitro and in vivo. Mechanistically, 6ME inhibited fatty acid oxidation (FAO) by binding to and decreasing the activity of PPARδ, a transcription factor involved in FAO.

Methods: AML cell lines and patient-derived cells were used to assess the activity of 6ME in vitro and in vivo. Computational methods, immunoblotting, and co-IP-HPLC analysis assessed the molecular target, and cellular consequence of 6ME activity.

Results: 6ME induced cytotoxicity of AML cell lines (IC50: 1.0 ± 0.13 μM) and patient-derived cells while sparing normal hematopoietic cells. Mouse engraftment studies showed that 6ME (5 mg/kg, three times/week for 4 weeks) selectively reduced patient-derived AML cell engraftment without affecting hematopoietic cell engraftment or imparting toxicity. Mechanistically, 6ME bound to and inhibited PPARδ leading to downregulated FAO gene expression (i.e., CD36 and CPT2) and reduced fatty acid cellular uptake resulting in FAO inhibition.

Conclusion: Pharmacological inhibition of PPARδ with 6ME is a novel approach to inducing selective death in AML.

背景:急性髓性白血病(AML)是一种破坏性的血液系统恶性肿瘤,治疗选择有限,生存预后差。因此,需要开发新的和选择性的抗aml治疗方法。6-甲氧基二氢鸟碱(6ME)是一种苯并苯胺生物碱,在体外和体内均可导致AML细胞选择性死亡。在机制上,6ME通过结合并降低PPARδ(一种参与FAO的转录因子)的活性来抑制脂肪酸氧化(FAO)。方法:采用AML细胞系和患者源性细胞体外和体内评价6ME的活性。计算方法、免疫印迹和协同ip - hplc分析评估了6ME活性的分子靶点和细胞后果。结果:6ME对AML细胞系和患者源性细胞具有细胞毒性(IC50: 1.0±0.13 μM),对正常造血细胞无影响。小鼠移植研究表明,6ME (5mg /kg,每周3次,连续4周)选择性地减少患者来源的AML细胞植入,而不影响造血细胞植入或增加毒性。从机制上讲,6ME结合并抑制PPARδ导致FAO基因表达下调(即CD36和CPT2),减少脂肪酸细胞摄取,导致FAO抑制。结论:6ME药物抑制PPARδ是一种诱导AML选择性死亡的新途径。
{"title":"Pharmacological inhibition of Peroxisome Proliferation-Activated Receptor Delta (PPARδ) imparts selective leukemia cell death.","authors":"Yingying Yang, Ekaterina Parfenova, Nikolina Vrdoljak, Mark Minden, Jessica Luc, Andrew C Doxey, Paul A Spagnuolo","doi":"10.1186/s40170-025-00402-5","DOIUrl":"10.1186/s40170-025-00402-5","url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML) is a devastating hematological malignancy with limited therapeutic options and poor survival outcomes. Therefore, the development of novel and selective anti-AML therapies is needed. 6-methoxydihydroavicine (6ME), a benzophenanthridine alkaloid, imparted selective AML cell death in vitro and in vivo. Mechanistically, 6ME inhibited fatty acid oxidation (FAO) by binding to and decreasing the activity of PPARδ, a transcription factor involved in FAO.</p><p><strong>Methods: </strong>AML cell lines and patient-derived cells were used to assess the activity of 6ME in vitro and in vivo. Computational methods, immunoblotting, and co-IP-HPLC analysis assessed the molecular target, and cellular consequence of 6ME activity.</p><p><strong>Results: </strong>6ME induced cytotoxicity of AML cell lines (IC<sub>50</sub>: 1.0 ± 0.13 μM) and patient-derived cells while sparing normal hematopoietic cells. Mouse engraftment studies showed that 6ME (5 mg/kg, three times/week for 4 weeks) selectively reduced patient-derived AML cell engraftment without affecting hematopoietic cell engraftment or imparting toxicity. Mechanistically, 6ME bound to and inhibited PPARδ leading to downregulated FAO gene expression (i.e., CD36 and CPT2) and reduced fatty acid cellular uptake resulting in FAO inhibition.</p><p><strong>Conclusion: </strong>Pharmacological inhibition of PPARδ with 6ME is a novel approach to inducing selective death in AML.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"36"},"PeriodicalIF":5.3,"publicationDate":"2025-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12297851/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144717610","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
Fatty acid synthase-derived lipid stores support breast cancer metastasis. 脂肪酸合酶衍生的脂质储存支持乳腺癌转移。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-07-10 DOI: 10.1186/s40170-025-00404-3
Chaylen Andolino, Eylem Kulkoyluoglu Cotul, Zilin Xianyu, Yun Li, Divya Bhat, Mitchell Ayers, Kimberly K Buhman, Stephen D Hursting, Michael K Wendt, Dorothy Teegarden

Lipid accumulation is associated with breast cancer metastasis. However, the mechanisms underlying how breast cancer cells increase lipid stores and their functional role in disease progression remain incompletely understood. Herein we quantified changes in lipid metabolism and characterized cytoplasmic lipid droplets in metastatic versus non-metastatic breast cancer cells. 14C-labeled palmitate was used to determine differences in fatty acid (FA) uptake and oxidation. Despite similar levels of palmitate uptake, metastatic cells increase lipid accumulation and oxidation of endogenous FAs compared to non-metastatic cells. Isotope tracing also demonstrated that metastatic cells support increased de novo lipogenesis by converting higher levels of glutamine and glucose into the FA precursor, citrate. Consistent with this, metastatic cells displayed increased levels of fatty acid synthase (FASN) and de novo lipogenesis. Genetic depletion or pharmacologic inhibition of FASN reduced cell migration, survival in anoikis assays, and in vivo metastasis. Finally, global proteomic analysis indicated that proteins involved in proteasome function, mitotic cell cycle, and intracellular protein transport were reduced following FASN inhibition of metastatic cells. Overall, these studies demonstrate that breast cancer metastases accumulate FAs by increasingde novo lipogenesis, storing TAG as cytoplasmic lipid droplets, and catabolizing these stores to drive several FAO-dependent steps in metastasis.

脂质积累与乳腺癌转移有关。然而,乳腺癌细胞增加脂质储存的机制及其在疾病进展中的功能作用仍不完全清楚。在这里,我们量化了脂质代谢的变化,并表征了转移性乳腺癌细胞与非转移性乳腺癌细胞的细胞质脂滴。14c标记棕榈酸酯用于测定脂肪酸(FA)摄取和氧化的差异。尽管棕榈酸摄取水平相似,但与非转移细胞相比,转移细胞增加了脂质积累和内源性FAs的氧化。同位素示踪还表明,转移细胞通过将较高水平的谷氨酰胺和葡萄糖转化为FA前体柠檬酸盐,支持增加的新生脂肪生成。与此一致的是,转移细胞显示出脂肪酸合成酶(FASN)和新生脂肪生成水平的增加。基因缺失或药物抑制FASN降低了细胞迁移,在anoikis试验中的存活和体内转移。最后,全球蛋白质组学分析表明,FASN抑制转移细胞后,参与蛋白酶体功能、有丝分裂细胞周期和细胞内蛋白质运输的蛋白质减少。总的来说,这些研究表明,乳腺癌转移通过增加新生脂肪生成,将TAG储存为细胞质脂滴,并分解这些储存来驱动转移中的几个依赖fao的步骤来积累FAs。
{"title":"Fatty acid synthase-derived lipid stores support breast cancer metastasis.","authors":"Chaylen Andolino, Eylem Kulkoyluoglu Cotul, Zilin Xianyu, Yun Li, Divya Bhat, Mitchell Ayers, Kimberly K Buhman, Stephen D Hursting, Michael K Wendt, Dorothy Teegarden","doi":"10.1186/s40170-025-00404-3","DOIUrl":"10.1186/s40170-025-00404-3","url":null,"abstract":"<p><p>Lipid accumulation is associated with breast cancer metastasis. However, the mechanisms underlying how breast cancer cells increase lipid stores and their functional role in disease progression remain incompletely understood. Herein we quantified changes in lipid metabolism and characterized cytoplasmic lipid droplets in metastatic versus non-metastatic breast cancer cells. <sup>14</sup>C-labeled palmitate was used to determine differences in fatty acid (FA) uptake and oxidation. Despite similar levels of palmitate uptake, metastatic cells increase lipid accumulation and oxidation of endogenous FAs compared to non-metastatic cells. Isotope tracing also demonstrated that metastatic cells support increased de novo lipogenesis by converting higher levels of glutamine and glucose into the FA precursor, citrate. Consistent with this, metastatic cells displayed increased levels of fatty acid synthase (FASN) and de novo lipogenesis. Genetic depletion or pharmacologic inhibition of FASN reduced cell migration, survival in anoikis assays, and in vivo metastasis. Finally, global proteomic analysis indicated that proteins involved in proteasome function, mitotic cell cycle, and intracellular protein transport were reduced following FASN inhibition of metastatic cells. Overall, these studies demonstrate that breast cancer metastases accumulate FAs by increasingde novo lipogenesis, storing TAG as cytoplasmic lipid droplets, and catabolizing these stores to drive several FAO-dependent steps in metastasis.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"35"},"PeriodicalIF":6.0,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12247306/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144607442","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
The role of B12 deficiency and methionine synthase in methionine-dependent cancer cells. B12缺乏和蛋氨酸合成酶在蛋氨酸依赖性癌细胞中的作用。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-07-02 DOI: 10.1186/s40170-025-00405-2
Mohamed M A El Husseiny, Roland Nilsson

Background: Human cells can synthesize methionine from homocysteine and folate-coupled methyl groups via the B12-dependent enzyme methionine synthase (MTR). Yet, it has been known for decades that cancer cells fail to grow when methionine is replaced by homocysteine, a phenomenon known as methionine dependence. The underlying mechanism remains unknown.

Methods: Cancer cell lines were cultured with homocysteine in place of methionine, and growth responses were measured. Revertant cells capable of growing in homocysteine were generated through long-term culture with high B12 and analyzed using single-cell RNA-seq. Metabolite uptake/release was measured using isotope dilution and MTR activity was assessed using metabolic flux analysis (MFA). Functional rescue experiments were performed by overexpressing the B12-independent methionine synthase enzyme.

Results: We report evidence that methionine dependence is caused by low MTR activity secondary to a B12 deficiency. High levels of the B12 cofactor were required to revert methionine-dependent cancer cells to grow on homocysteine. The adapted "revertant" cells display gene expression signatures consistent with reduced invasion and metastasis. Metabolic flux analysis indicated that methionine-dependent cells do not fully activate MTR when cultured in homocysteine. High concentrations of homocysteine partially rescued growth of methionine-dependent cells. Expression of a B12-independent methionine synthase enzyme in cancer cells restored growth on homocysteine and normalized the SAM:SAH ratio, while overexpression of the B12-dependent human enzyme had no effect.

Conclusion: Methionine dependence in cancer can be driven by low MTR activity secondary to B12 deficiency, at least in the cell lines studied. This mechanistic insight resolves a long-standing question in cancer metabolism and may open new avenues for exploiting the phenomenon for cancer therapy.

背景:人类细胞可以通过b12依赖的蛋氨酸合成酶(MTR)从同型半胱氨酸和叶酸偶联的甲基合成蛋氨酸。然而,几十年来人们已经知道,当蛋氨酸被同型半胱氨酸取代时,癌细胞无法生长,这种现象被称为蛋氨酸依赖。其潜在机制尚不清楚。方法:用同型半胱氨酸代替蛋氨酸培养癌细胞,观察其生长反应。通过高B12长期培养产生能在同型半胱氨酸中生长的逆转细胞,并使用单细胞RNA-seq进行分析。用同位素稀释法测定代谢物的吸收/释放,用代谢通量分析(MFA)评估MTR活性。通过过表达b12独立的蛋氨酸合成酶进行功能拯救实验。结果:我们报告的证据表明,蛋氨酸依赖是由继发于B12缺乏的低MTR活性引起的。需要高水平的B12辅助因子来恢复依赖蛋氨酸的癌细胞在同型半胱氨酸上的生长。适应的“逆转录”细胞显示出与减少侵袭和转移相一致的基因表达特征。代谢通量分析表明,蛋氨酸依赖细胞在同型半胱氨酸中培养时不能完全激活MTR。高浓度的同型半胱氨酸部分地挽救了蛋氨酸依赖细胞的生长。在癌细胞中表达一种不依赖于b12的蛋氨酸合成酶可以恢复同型半胱氨酸的生长,并使SAM:SAH比例正常化,而过度表达这种依赖于b12的人酶则没有影响。结论:至少在所研究的细胞系中,癌症对蛋氨酸的依赖可能是由继发于B12缺乏的低MTR活性驱动的。这种机制的见解解决了癌症代谢的一个长期存在的问题,并可能为利用这种现象进行癌症治疗开辟新的途径。
{"title":"The role of B<sub>12</sub> deficiency and methionine synthase in methionine-dependent cancer cells.","authors":"Mohamed M A El Husseiny, Roland Nilsson","doi":"10.1186/s40170-025-00405-2","DOIUrl":"10.1186/s40170-025-00405-2","url":null,"abstract":"<p><strong>Background: </strong>Human cells can synthesize methionine from homocysteine and folate-coupled methyl groups via the B<sub>12</sub>-dependent enzyme methionine synthase (MTR). Yet, it has been known for decades that cancer cells fail to grow when methionine is replaced by homocysteine, a phenomenon known as methionine dependence. The underlying mechanism remains unknown.</p><p><strong>Methods: </strong>Cancer cell lines were cultured with homocysteine in place of methionine, and growth responses were measured. Revertant cells capable of growing in homocysteine were generated through long-term culture with high B<sub>12</sub> and analyzed using single-cell RNA-seq. Metabolite uptake/release was measured using isotope dilution and MTR activity was assessed using metabolic flux analysis (MFA). Functional rescue experiments were performed by overexpressing the B<sub>12</sub>-independent methionine synthase enzyme.</p><p><strong>Results: </strong>We report evidence that methionine dependence is caused by low MTR activity secondary to a B<sub>12</sub> deficiency. High levels of the B<sub>12</sub> cofactor were required to revert methionine-dependent cancer cells to grow on homocysteine. The adapted \"revertant\" cells display gene expression signatures consistent with reduced invasion and metastasis. Metabolic flux analysis indicated that methionine-dependent cells do not fully activate MTR when cultured in homocysteine. High concentrations of homocysteine partially rescued growth of methionine-dependent cells. Expression of a B<sub>12</sub>-independent methionine synthase enzyme in cancer cells restored growth on homocysteine and normalized the SAM:SAH ratio, while overexpression of the B<sub>12</sub>-dependent human enzyme had no effect.</p><p><strong>Conclusion: </strong>Methionine dependence in cancer can be driven by low MTR activity secondary to B<sub>12</sub> deficiency, at least in the cell lines studied. This mechanistic insight resolves a long-standing question in cancer metabolism and may open new avenues for exploiting the phenomenon for cancer therapy.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"34"},"PeriodicalIF":6.0,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12220533/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144552385","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
Dexmedetomidine promotes colorectal cancer progression mediated by gamma-aminobutyric acid signaling. 右美托咪定促进γ -氨基丁酸信号介导的结直肠癌进展。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-06-23 DOI: 10.1186/s40170-025-00403-4
Jing Dong, Yuanyuan Wu, Ji Che, Zhiyong He, Jun Zhang
{"title":"Dexmedetomidine promotes colorectal cancer progression mediated by gamma-aminobutyric acid signaling.","authors":"Jing Dong, Yuanyuan Wu, Ji Che, Zhiyong He, Jun Zhang","doi":"10.1186/s40170-025-00403-4","DOIUrl":"10.1186/s40170-025-00403-4","url":null,"abstract":"","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"33"},"PeriodicalIF":6.0,"publicationDate":"2025-06-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12186342/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144473976","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
Obesity and cervical intraepithelial neoplasia: regulation of mitochondrial energy metabolism via the Kisspeptin/GPR54 signaling pathway. 肥胖和宫颈上皮内瘤变:通过Kisspeptin/GPR54信号通路调节线粒体能量代谢
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-06-18 DOI: 10.1186/s40170-025-00398-y
Jiajia Pan, Yuanyuan Chen, Yan Ye, Peipei Li, Feifei Ni, Haizhen He

Background: Obesity exacerbates the severity of cervical intraepithelial neoplasia (CIN), potentially through metabolic alterations. This study investigates how the Kisspeptin/GPR54 signaling pathway mediates mitochondrial energy metabolism in obesity-related CIN.

Methods: A clinical analysis of 980 samples was conducted to assess the correlation between Body Mass Index (BMI) and CIN grade. Transcriptomic analysis identified KISS1R as a key gene. Functional assays in cervical cancer (CC) cell lines, including CCK-8, wound healing, and Transwell assays, were used to evaluate the effects of KISS1 modulation. Mitochondrial function was assessed via oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) assays. A high-fat diet-induced CIN mouse model was used to investigate the in vivo effects.

Results: BMI positively correlated with CIN grade, with elevated KISS1R expression in higher CIN grades. Overexpression of KISS1 enhanced CC cell proliferation and migration by reprogramming mitochondrial energy metabolism. In high-fat environments, KISS1 silencing and mitochondrial activator PQQ modulated CC cell behavior. Activation of Kisspeptin/GPR54 in obese CIN mice exacerbated cervical lesions.

Conclusion: The Kisspeptin/GPR54 signaling pathway enhances mitochondrial energy metabolism, promoting obesity-related CIN grade. These findings provide a potential molecular mechanism linking obesity to CC and suggest new therapeutic targets.

背景:肥胖可能通过代谢改变加重宫颈上皮内瘤变(CIN)的严重程度。本研究探讨Kisspeptin/GPR54信号通路如何介导肥胖相关CIN的线粒体能量代谢。方法:对980例患者进行临床分析,评价身体质量指数(BMI)与CIN分级的相关性。转录组学分析鉴定KISS1R为关键基因。宫颈癌细胞系的功能测定,包括CCK-8、伤口愈合和Transwell测定,用于评估KISS1调节的作用。通过耗氧率(OCR)和细胞外酸化率(ECAR)测定评估线粒体功能。采用高脂饮食诱导的CIN小鼠模型研究其体内效应。结果:BMI与CIN分级呈正相关,较高CIN分级KISS1R表达升高。KISS1过表达通过重编程线粒体能量代谢增强CC细胞增殖和迁移。在高脂肪环境中,KISS1沉默和线粒体激活因子PQQ调节CC细胞的行为。Kisspeptin/GPR54在肥胖CIN小鼠中的激活加重了宫颈病变。结论:Kisspeptin/GPR54信号通路增强线粒体能量代谢,促进肥胖相关CIN分级。这些发现提供了将肥胖与CC联系起来的潜在分子机制,并提出了新的治疗靶点。
{"title":"Obesity and cervical intraepithelial neoplasia: regulation of mitochondrial energy metabolism via the Kisspeptin/GPR54 signaling pathway.","authors":"Jiajia Pan, Yuanyuan Chen, Yan Ye, Peipei Li, Feifei Ni, Haizhen He","doi":"10.1186/s40170-025-00398-y","DOIUrl":"10.1186/s40170-025-00398-y","url":null,"abstract":"<p><strong>Background: </strong>Obesity exacerbates the severity of cervical intraepithelial neoplasia (CIN), potentially through metabolic alterations. This study investigates how the Kisspeptin/GPR54 signaling pathway mediates mitochondrial energy metabolism in obesity-related CIN.</p><p><strong>Methods: </strong>A clinical analysis of 980 samples was conducted to assess the correlation between Body Mass Index (BMI) and CIN grade. Transcriptomic analysis identified KISS1R as a key gene. Functional assays in cervical cancer (CC) cell lines, including CCK-8, wound healing, and Transwell assays, were used to evaluate the effects of KISS1 modulation. Mitochondrial function was assessed via oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) assays. A high-fat diet-induced CIN mouse model was used to investigate the in vivo effects.</p><p><strong>Results: </strong>BMI positively correlated with CIN grade, with elevated KISS1R expression in higher CIN grades. Overexpression of KISS1 enhanced CC cell proliferation and migration by reprogramming mitochondrial energy metabolism. In high-fat environments, KISS1 silencing and mitochondrial activator PQQ modulated CC cell behavior. Activation of Kisspeptin/GPR54 in obese CIN mice exacerbated cervical lesions.</p><p><strong>Conclusion: </strong>The Kisspeptin/GPR54 signaling pathway enhances mitochondrial energy metabolism, promoting obesity-related CIN grade. These findings provide a potential molecular mechanism linking obesity to CC and suggest new therapeutic targets.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"31"},"PeriodicalIF":6.0,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175464/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144324570","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
NRF2 maintains redox balance via ME1 and NRF2 inhibitor synergizes with venetoclax in NPM1-mutated acute myeloid leukemia. NRF2通过ME1维持氧化还原平衡,NRF2抑制剂在npm1突变的急性髓性白血病中与venetoclax协同作用。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-06-18 DOI: 10.1186/s40170-025-00401-6
Jiayuan Hu, Zihao Yuan, Yan Shu, Jun Ren, Jing Yang, Lisha Tang, Xingyu Wei, Yongcan Liu, Fangfang Jin, Qiaoling Xiao, Xinyi Chen, Nan Wu, Wen Zhao, Ziwei Li, Ling Zhang

Background: Acute myeloid leukemia (AML) with nucleophosmin 1 (NPM1) mutations represents a distinct subtype of leukemia. Emerging evidence suggests that regulation of redox metabolism contributes to tumorigenesis and reveals a metabolic vulnerability in anti-tumor therapies. However, the role of redox homeostasis between reactive oxygen species (ROS) and antioxidant systems plays in NPM1-mutated AML has not been fully elucidated.

Methods: First, ROS-related metabolic pathways in NPM1-mutated AML were analyzed using RNA-sequencing data. Intracellular and mitochondrial ROS levels in leukemia cells were detected using flow cytometry (FCM). The expression of nuclear factor (erythroid-derived 2)-like 2 (NRF2) was analyzed in public databases and further validated in AML primary blasts and cell lines by quantitative real-time PCR (qRT-PCR), western blotting, and immunofluorescence. Next, the mechanism underlying NRF2 expression was investigated through the RNA immunoprecipitation (RIP), methylated RNA immunoprecipitation (MeRIP) and rescue experiments. Additionally, the downstream target gene of NRF2 was identified by bioinformatics analysis and chromatin immunoprecipitation (ChIP) assays. Furthermore, RNA interference and the NRF2 inhibitor ML385 were applied to explore the role of NRF2 in leukemia. Finally, the anti-leukemic effects of ML385 alone or in combination with the B-cell lymphoma 2 (BCL-2) inhibitor venetoclax on AML cells were investigated using FCM analysis and western blotting, and further explored in cell line-derived xenograft (CDX) mouse models.

Results: In this study, we identified significant ROS accumulation in leukemia cells with NPM1 mutations. Meanwhile, elevated NRF2 expression and its nuclear localization were observed in NPM1-mutated AML cells. The high NRF2 expression levels were at least partially induced by fat mass and obesity-associated protein (FTO) via m6A modification. Functionally, NRF2 exerts its antioxidant effects by transcriptionally upregulating malic enzyme 1 (ME1) expression and enhancing its activity. Targeting NRF2/ME1 axis reduced NADPH/NADP+ ratio, increased ROS levels, impaired leukemia cell viability, and promoted apoptosis. More importantly, NRF2 inhibitor ML385 in combination with venetoclax showed synergistic anti-leukemic activity in vitro and in vivo.

Conclusion: Overall, our findings provide new insight into the therapeutic potential of targeting NRF2 and guide the development of innovative combination therapies in NPM1-mutated AML.

背景:核磷蛋白1 (NPM1)突变的急性髓性白血病(AML)是一种独特的白血病亚型。新出现的证据表明,氧化还原代谢的调节有助于肿瘤的发生,并揭示了抗肿瘤治疗中的代谢脆弱性。然而,活性氧(ROS)和抗氧化系统之间的氧化还原稳态在npm1突变的AML中所起的作用尚未完全阐明。方法:首先,利用rna测序数据分析npm1突变AML中ros相关代谢途径。采用流式细胞术(FCM)检测白血病细胞内及线粒体ROS水平。在公共数据库中分析核因子(红细胞衍生2)样2 (NRF2)的表达,并通过实时荧光定量PCR (qRT-PCR)、western blotting和免疫荧光进一步验证其在AML原代细胞和细胞系中的表达。接下来,我们通过RNA免疫沉淀(RIP)、甲基化RNA免疫沉淀(MeRIP)和抢救实验来研究NRF2表达的机制。此外,通过生物信息学分析和染色质免疫沉淀(ChIP)试验鉴定了NRF2的下游靶基因。此外,我们还利用RNA干扰和NRF2抑制剂ML385来探讨NRF2在白血病中的作用。最后,通过流式细胞术分析和western blotting研究ML385单独或联合b细胞淋巴瘤2 (BCL-2)抑制剂venetoclax对AML细胞的抗白血病作用,并在细胞系来源的异种移植(CDX)小鼠模型中进一步探索。结果:在本研究中,我们在NPM1突变的白血病细胞中发现了明显的ROS积累。同时,在npm1突变的AML细胞中,NRF2的表达和核定位升高。NRF2的高表达水平至少部分是由脂肪量和肥胖相关蛋白(FTO)通过m6A修饰诱导的。在功能上,NRF2通过上调苹果酸酶1 (ME1)的转录表达并增强其活性来发挥抗氧化作用。靶向NRF2/ME1轴可降低NADPH/NADP+比值,提高ROS水平,损害白血病细胞活力,促进细胞凋亡。更重要的是,NRF2抑制剂ML385与venetoclax联合在体外和体内均显示出协同抗白血病活性。结论:总的来说,我们的研究结果为靶向NRF2的治疗潜力提供了新的见解,并指导了npm1突变AML的创新联合治疗的发展。
{"title":"NRF2 maintains redox balance via ME1 and NRF2 inhibitor synergizes with venetoclax in NPM1-mutated acute myeloid leukemia.","authors":"Jiayuan Hu, Zihao Yuan, Yan Shu, Jun Ren, Jing Yang, Lisha Tang, Xingyu Wei, Yongcan Liu, Fangfang Jin, Qiaoling Xiao, Xinyi Chen, Nan Wu, Wen Zhao, Ziwei Li, Ling Zhang","doi":"10.1186/s40170-025-00401-6","DOIUrl":"10.1186/s40170-025-00401-6","url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML) with nucleophosmin 1 (NPM1) mutations represents a distinct subtype of leukemia. Emerging evidence suggests that regulation of redox metabolism contributes to tumorigenesis and reveals a metabolic vulnerability in anti-tumor therapies. However, the role of redox homeostasis between reactive oxygen species (ROS) and antioxidant systems plays in NPM1-mutated AML has not been fully elucidated.</p><p><strong>Methods: </strong>First, ROS-related metabolic pathways in NPM1-mutated AML were analyzed using RNA-sequencing data. Intracellular and mitochondrial ROS levels in leukemia cells were detected using flow cytometry (FCM). The expression of nuclear factor (erythroid-derived 2)-like 2 (NRF2) was analyzed in public databases and further validated in AML primary blasts and cell lines by quantitative real-time PCR (qRT-PCR), western blotting, and immunofluorescence. Next, the mechanism underlying NRF2 expression was investigated through the RNA immunoprecipitation (RIP), methylated RNA immunoprecipitation (MeRIP) and rescue experiments. Additionally, the downstream target gene of NRF2 was identified by bioinformatics analysis and chromatin immunoprecipitation (ChIP) assays. Furthermore, RNA interference and the NRF2 inhibitor ML385 were applied to explore the role of NRF2 in leukemia. Finally, the anti-leukemic effects of ML385 alone or in combination with the B-cell lymphoma 2 (BCL-2) inhibitor venetoclax on AML cells were investigated using FCM analysis and western blotting, and further explored in cell line-derived xenograft (CDX) mouse models.</p><p><strong>Results: </strong>In this study, we identified significant ROS accumulation in leukemia cells with NPM1 mutations. Meanwhile, elevated NRF2 expression and its nuclear localization were observed in NPM1-mutated AML cells. The high NRF2 expression levels were at least partially induced by fat mass and obesity-associated protein (FTO) via m<sup>6</sup>A modification. Functionally, NRF2 exerts its antioxidant effects by transcriptionally upregulating malic enzyme 1 (ME1) expression and enhancing its activity. Targeting NRF2/ME1 axis reduced NADPH/NADP<sup>+</sup> ratio, increased ROS levels, impaired leukemia cell viability, and promoted apoptosis. More importantly, NRF2 inhibitor ML385 in combination with venetoclax showed synergistic anti-leukemic activity in vitro and in vivo.</p><p><strong>Conclusion: </strong>Overall, our findings provide new insight into the therapeutic potential of targeting NRF2 and guide the development of innovative combination therapies in NPM1-mutated AML.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"32"},"PeriodicalIF":6.0,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12177962/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144324569","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
Coordinated histone methylation loss and MYC activation promote translational capacity under amino acid restriction. 协同组蛋白甲基化丢失和MYC激活促进氨基酸限制下的翻译能力。
IF 6 3区 医学 Q1 CELL BIOLOGY Pub Date : 2025-06-16 DOI: 10.1186/s40170-025-00399-x
Chen Cheng, Trent Su, Marco Morselli, Siavash K Kurdistani

Background: Cells adapt to nutrient fluctuations through both signaling and epigenetic mechanisms. While amino acid (AA) deprivation is known to suppress protein synthesis via mTORC1 inactivation, the epigenetic pathways that support cellular adaptation and recovery remain poorly understood. We investigated how chromatin and transcriptional changes contribute to maintaining translational capacity during AA restriction and priming cells for growth upon AA repletion.

Methods: Human cells were cultured under amino acid-replete or -depleted conditions, and global histone methylation levels were assessed by Western blotting and ChIP-seq. RNA-seq and chromatin-associated RNA-seq (chromRNA-seq) were used to evaluate gene expression and transcriptional output. Ribosome profiling and [35S]-methionine/cysteine or O-propargyl-puromycin (OPP) incorporation assays measured protein synthesis. Functional contributions of SETD8 and MYC were tested through knockdown and overexpression experiments.

Results: AA deprivation induced a selective, genome-wide loss of H4K20me1, particularly from gene bodies, and led to increased MYC expression and binding at promoter regions. These changes were most pronounced at genes encoding ribosomal proteins and translation initiation factors. Although overall protein synthesis declined during AA restriction, these cells showed increased translational capacity evidenced by accumulation of monomeric ribosomes and enhanced translation upon AA repletion. Loss of H4K20me1 was independent of mTORC1 signaling and partly driven by SETD8 protein downregulation. While MYC overexpression alone was insufficient to upregulate translation-related genes, its combination with SETD8 knockdown in nutrient-rich conditions was both necessary and sufficient to induce expression of these genes and enhance protein synthesis.

Conclusions: Our findings reveal a chromatin-based mechanism by which cells integrate metabolic status with transcriptional regulation to adapt to amino acid limitation. Loss of H4K20me1 and increased MYC activity act in parallel to prime the translational machinery during AA deprivation, enabling rapid recovery of protein synthesis upon nutrient restoration. This mechanism may help explain how cells maintain competitive growth potential under fluctuating nutrient conditions and has implications for understanding MYC-driven cancer progression.

背景:细胞通过信号和表观遗传机制适应营养波动。虽然已知氨基酸(AA)剥夺通过mTORC1失活抑制蛋白质合成,但支持细胞适应和恢复的表观遗传途径仍然知之甚少。我们研究了染色质和转录变化如何在AA限制期间维持翻译能力,以及在AA补充时启动细胞生长。方法:将人细胞在氨基酸充满或缺失的条件下培养,采用Western blotting和ChIP-seq技术检测组蛋白甲基化水平。RNA-seq和染色质相关RNA-seq (chromRNA-seq)用于评估基因表达和转录输出。核糖体分析和[35S]-蛋氨酸/半胱氨酸或o -丙炔-嘌呤霉素(OPP)掺入试验测量蛋白质合成。通过敲低和过表达实验检测SETD8和MYC的功能贡献。结果:AA剥夺导致H4K20me1选择性全基因组缺失,尤其是基因体缺失,并导致MYC在启动子区域的表达和结合增加。这些变化在编码核糖体蛋白和翻译起始因子的基因上最为明显。虽然在AA限制期间整体蛋白质合成下降,但这些细胞的翻译能力增强,这可以通过单体核糖体的积累和AA补充后的翻译能力增强来证明。H4K20me1的缺失独立于mTORC1信号传导,部分由SETD8蛋白下调驱动。虽然MYC过表达不足以上调翻译相关基因,但在营养丰富的条件下,MYC与SETD8敲低的结合是诱导这些基因表达和增强蛋白质合成的必要和充分条件。结论:我们的研究结果揭示了一种基于染色质的机制,通过该机制,细胞将代谢状态与转录调节结合起来,以适应氨基酸的限制。在AA剥夺过程中,H4K20me1的缺失和MYC活性的增加同时启动了翻译机制,使营养恢复后蛋白质合成迅速恢复。这一机制可能有助于解释细胞如何在波动的营养条件下保持竞争性生长潜力,并有助于理解myc驱动的癌症进展。
{"title":"Coordinated histone methylation loss and MYC activation promote translational capacity under amino acid restriction.","authors":"Chen Cheng, Trent Su, Marco Morselli, Siavash K Kurdistani","doi":"10.1186/s40170-025-00399-x","DOIUrl":"10.1186/s40170-025-00399-x","url":null,"abstract":"<p><strong>Background: </strong>Cells adapt to nutrient fluctuations through both signaling and epigenetic mechanisms. While amino acid (AA) deprivation is known to suppress protein synthesis via mTORC1 inactivation, the epigenetic pathways that support cellular adaptation and recovery remain poorly understood. We investigated how chromatin and transcriptional changes contribute to maintaining translational capacity during AA restriction and priming cells for growth upon AA repletion.</p><p><strong>Methods: </strong>Human cells were cultured under amino acid-replete or -depleted conditions, and global histone methylation levels were assessed by Western blotting and ChIP-seq. RNA-seq and chromatin-associated RNA-seq (chromRNA-seq) were used to evaluate gene expression and transcriptional output. Ribosome profiling and [<sup>35</sup>S]-methionine/cysteine or O-propargyl-puromycin (OPP) incorporation assays measured protein synthesis. Functional contributions of SETD8 and MYC were tested through knockdown and overexpression experiments.</p><p><strong>Results: </strong>AA deprivation induced a selective, genome-wide loss of H4K20me1, particularly from gene bodies, and led to increased MYC expression and binding at promoter regions. These changes were most pronounced at genes encoding ribosomal proteins and translation initiation factors. Although overall protein synthesis declined during AA restriction, these cells showed increased translational capacity evidenced by accumulation of monomeric ribosomes and enhanced translation upon AA repletion. Loss of H4K20me1 was independent of mTORC1 signaling and partly driven by SETD8 protein downregulation. While MYC overexpression alone was insufficient to upregulate translation-related genes, its combination with SETD8 knockdown in nutrient-rich conditions was both necessary and sufficient to induce expression of these genes and enhance protein synthesis.</p><p><strong>Conclusions: </strong>Our findings reveal a chromatin-based mechanism by which cells integrate metabolic status with transcriptional regulation to adapt to amino acid limitation. Loss of H4K20me1 and increased MYC activity act in parallel to prime the translational machinery during AA deprivation, enabling rapid recovery of protein synthesis upon nutrient restoration. This mechanism may help explain how cells maintain competitive growth potential under fluctuating nutrient conditions and has implications for understanding MYC-driven cancer progression.</p>","PeriodicalId":9418,"journal":{"name":"Cancer & Metabolism","volume":"13 1","pages":"29"},"PeriodicalIF":6.0,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168343/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144309573","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:604180095
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