11,002 results on '"Glutamine metabolism"'
Search Results
2. RUNX2 enhances bladder cancer progression by promoting glutamine metabolism
- Author
-
Huang, Zhigang, Liu, Bin, Li, Xiaoju, Jin, Chenghua, Hu, Quansen, Zhao, Zhiwei, Sun, Yimin, and Wang, Qian
- Published
- 2025
- Full Text
- View/download PDF
3. Glutamine metabolism is essential for coronavirus replication in host cells and in mice
- Author
-
Greene, Kai Su, Choi, Annette, Yang, Nianhui, Chen, Matthew, Li, Ruizhi, Qiu, Yijian, Ezzatpour, Shahrzad, Rojas, Katherine S., Shen, Jonathan, Wilson, Kristin F., Katt, William P., Aguilar, Hector C., Lukey, Michael J., Whittaker, Gary R., and Cerione, Richard A.
- Published
- 2025
- Full Text
- View/download PDF
4. Sono-mediated glutamine metabolic nanoplatform against liver fibrosis via breaking the vicious self-injury of activated hepatic stellate cells
- Author
-
Yang, Peng, Liu, Weijing, Chen, Zhuang, Duan, Deshang, Xu, Jiamin, An, Xian, Xie, Anna, Rao, Zhiping, Xia, Yuqiong, Zhang, Ruili, Ning, Pengbo, Qiao, Chaoqiang, Zhang, Xianghan, and Wang, Zhongliang
- Published
- 2025
- Full Text
- View/download PDF
5. Inhibition of glutaminase 1 reduces M1 macrophage polarization to protect against monocrotaline-induced pulmonary arterial hypertension
- Author
-
Chen, Xing, Li, Lixiang, Deng, Yan, Liao, Juan, Meng, Hui, Liang, Limei, Hu, Jie, Xie, Dongwei, and Liang, Guizi
- Published
- 2025
- Full Text
- View/download PDF
6. Folate-targeted nanoparticles for glutamine metabolism inhibition enhance anti-tumor immunity and suppress tumor growth in ovarian cancer
- Author
-
Chen, Shuning, Jiang, Yu, Zheng, Jiao, Li, Pan, Liu, Maoyu, Zhu, Yi, Zhu, Shenyin, and Chang, Shufang
- Published
- 2025
- Full Text
- View/download PDF
7. Identification of a novel prognostic model for gastric cancer utilizing glutamine-related genes
- Author
-
Li, Weidong, Zhong, Qixing, Deng, Naisheng, Wang, Haitao, Ouyang, Jun, Guan, Zhifen, Zhou, Xinhao, Li, Kai, Sun, Xueying, and Wang, Yao
- Published
- 2024
- Full Text
- View/download PDF
8. Mitochondria-anchoring self-assembled nanoparticles for multi-path energy depletion: A “nano bomb” in chemo-co-starvation therapy
- Author
-
Zhang, Kexin, Zhu, Jiaxin, Wang, Ruyi, Zhu, Wanfang, Zhang, Zhongtao, Gong, Liangping, Feng, Feng, Liu, Wenyuan, Han, Lingfei, and Qu, Wei
- Published
- 2023
- Full Text
- View/download PDF
9. Glutamine metabolism in breast cancer and possible therapeutic targets
- Author
-
Li, Shiqi, Zeng, Hui, Fan, Junli, Wang, Fubing, Xu, Chen, Li, Yirong, Tu, Jiancheng, Nephew, Kenneth P., and Long, Xinghua
- Published
- 2023
- Full Text
- View/download PDF
10. Glutamine and cancer: metabolism, immune microenvironment, and therapeutic targets.
- Author
-
Nan, Ding, Yao, Weiping, Huang, Luanluan, Liu, Ruiqi, Chen, Xiaoyan, Xia, Wenjie, Sheng, Hailong, Zhang, Haibo, Liang, Xiaodong, and Lu, Yanwei
- Subjects
- *
METABOLIC reprogramming , *DIETARY supplements , *TUMOR microenvironment , *CELL metabolism , *LIFE sciences - Abstract
Glutamine is the most abundant amino acid in human serum, and it can provide carbon and nitrogen for biosynthesis, which is crucial for proliferating cells. Moreover, it is widely known that glutamine metabolism is reprogrammed in cancer cells. Many cancer cells undergo metabolic reprogramming targeting glutamine, increasing its uptake to meet their rapid proliferation demands. An increasing amount of study is being done on the particular glutamine metabolic pathways in cancer cells. Further investigation into the function of glutamine in immune cells is warranted given the critical role these cells play in the fight against cancer. Immune cells use glutamine for a variety of biological purposes, including the growth, differentiation, and destruction of cancer cells. With the encouraging results of cancer immunotherapy in recent years, more investigation into the impact of glutamine metabolism on immune cell function in the cancer microenvironment could lead to the discovery of new targets and therapeutic approaches. Oral supplementation with glutamine also enhances the immune capabilities of cancer patients, improves the sensitivity to chemotherapy and radiotherapy, and improves prognosis. The unique metabolism of glutamine in cancer cells, its function in various immune cells, the impact of inhibitors of glutamine metabolism, and the therapeutic use of glutamine supplements are all covered in detail in this article. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. Infectious Spleen and Kidney Necrosis Virus ORF093R and ORF102R Regulate Glutamate Metabolic Reprogramming to Support Virus Proliferation by Interacting with c-Myc.
- Author
-
Niu, Yinjie, Ye, Caimei, Lin, Qiang, Liang, Hongru, Luo, Xia, Ma, Baofu, Li, Ningqiu, and Fu, Xiaozhe
- Subjects
- *
METABOLIC reprogramming , *GLUTAMATE dehydrogenase , *ISOCITRATE dehydrogenase , *GENE expression , *ENZYME metabolism , *GLUTAMINE synthetase - Abstract
Glutamine metabolism is essential for infectious spleen and kidney necrosis virus (ISKNV) replication. Glutaminase 1 (GLS1), the key enzyme of the glutamine metabolism, and c-Myc positively regulate ISKNV infection, while c-Myc is closely correlated with GLS1. However, the regulatory mechanism among ISKNV, c-Myc and glutamine metabolism remains unclear. Here, we indicated that c-Myc increased glutamine uptake by increasing the GLS1, glutamate dehydrogenase (GDH) and isocitrate dehydrogenase (IDH2) expression of glutamine metabolism. ISKNV ORF102R, ORF093R and ORF118L co-located with c-Myc in CPB cells. Co-IP results showed that ISKNV ORF102R and ORF093R interacted with c-Myc, while ORF118L did not interact with c-Myc. The expression levels of c-Myc, GLS1 and IDH2 were increased in ISKNV ORF093R expression cells, and the mRNA and protein levels of GLS1 were upregulated in ISKNV 102R-expressing cells. These results indicated that ISKNV reconstructed glutamine metabolism to satisfy the energy and macromolecule requirements for virus proliferation by ORF093R and ORF102R interacting with c-Myc, which provides the foundation for innovative antiviral strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
12. 长非编码RNA KCNQ1OT1 的泛癌分析及其在胃癌中对 谷氨酰胺代谢的调控作用.
- Author
-
于亚楠, 李家秋, and 马晓林
- Abstract
The long non-coding RNA KCNQ10T1 plays an important role in promoting the occurrence and development of various cancers. However, there is currently no systematic analysis of KCNQ10T1 in pan cancer. To elucidate the value of KCNQ10T1 in tumor diagnosis and prognosis, this study analyzed its expression levels in pan-cancer tissues and its impact on patient prognosis. By analyzing the regulatory mechanism of KCNQ10T1 in gastric cancer, new molecular targets may be found for the diagnosis and treatment of gastric cancer. Using Sangerbox 3.0, ACLBI and UALCAN databases, we found the expression levels of KCNQ10T1 were increased in 7 tumor tissues types (P<0.05). We found KCNQ10T1 expression was correlated with poor prognosis in many tumor types using Sangerbox 3. 0 database. We used R software to analyze the differential genes between the high and low expression groups of KCNQ10T1 in gastric cancer patients (P<0.05, log2 FoldChange>1). The GO and KEGG enrichment analysis showed that KCNQ10T1 was involved in the glutamine metabolism of gastric cancer. The cell counting and Western blot detection showed that knocking down KCNQ10T1 significantly reduced the gastric cancer cell activity, SLC1A5 expression level and SLC1A5-mediated glutamine transport process (P<0.01). Bioinformatics, RNA immunoprecipitation and dual luciferase analysis confirmed that KCNQ1OTI competitively bind to miR-138-5p to promote the expression of SLC1A5. Finally, ChIP-seq data was used to detect the high H3K27ac signaling at the gene locus of KCNQ1OT1, and ChIP-qPCR was used to verify that P300-mediated enhancer activity regulated the high expression of KCNQ10T1 in gastric cancer. KC- NQ1OTI can serve as an independent diagnostic biomarker and prognostic predictor in various tumors. Targeting the KCNQ1OT1/miR-138-5p/SLC1A5 signaling axis to regulate glutamine metabolism may provide new strategies and molecular targets for the treatment of gastric cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
13. Metabolic-Modulating Effects of Radiation: Undetectable Yet Deadly—A Review on Radiotherapy.
- Author
-
Fiorica, Francesco, Tebano, Umberto, Napoli, Giuseppe, Franceschetto, Antonella, Muraro, Marco, Giorgi, Carlotta, and Pinton, Paolo
- Subjects
- *
ARGININE metabolism , *AMINO acid metabolism , *GLUTAMINE metabolism , *LIPID metabolism , *GLYCINE metabolism , *SERINE metabolism , *RADIOTHERAPY , *MITOCHONDRIA , *METABOLIC reprogramming , *REACTIVE oxygen species , *METABOLISM , *TUMORS , *METABOLOMICS , *RADIATION doses , *FATTY acids , *NANOPARTICLES , *GLYCOSAMINOGLYCANS , *WARBURG Effect (Oncology) - Abstract
Simple Summary: This review explores how radiotherapy disrupts critical metabolic pathways in cancer cells. It highlights vulnerabilities in glucose metabolism, lipid synthesis, and amino acid pathways, which radiotherapy exploits to enhance tumour control. Advancements such as nanoparticle-enhanced and FLASH radiotherapy are discussed for their ability to improve precision and reduce damage to healthy tissues. Tools like FDG-PET scans are emphasised for detecting metabolic changes and guiding therapy combinations, such as precision radiotherapy with metabolic inhibitors. This review also outlines future prospects, including personalised metabolic imaging, integration with systemic therapies, targeting tumour-specific metabolic dependencies, and innovations like immune modulation and ferroptosis induction. These approaches aim to transform radiotherapy from a localised treatment to a systemic strategy that targets cancer's metabolic complexities, improving both efficacy and patient outcomes. Radiotherapy has traditionally been viewed as a localised treatment modality, primarily inducing DNA damage to eradicate cancer cells. However, emerging research reveals that radiation also profoundly affects cellular metabolism, particularly in reprogramming tumour cells' energy production and survival pathways. This article explores the role of metabolic reprogramming in the response of cancer cells to radiotherapy, highlighting how these biological processes can be leveraged to enhance treatment efficacy and overcome resistance. Radiotherapy is also discussed, focusing on how metabolic and molecular profiling of tumours can help personalise radiation treatments to improve outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
14. Targeting Asparagine Metabolism in Solid Tumors.
- Author
-
Hanada, Keita, Kawada, Kenji, and Obama, Kazutaka
- Abstract
Reprogramming of energy metabolism to support cellular growth is a "hallmark" of cancer, allowing cancer cells to balance the catabolic demands with the anabolic needs of producing the nucleotides, amino acids, and lipids necessary for tumor growth. Metabolic alterations, or "addiction", are promising therapeutic targets and the focus of many drug discovery programs. Asparagine metabolism has gained much attention in recent years as a novel target for cancer therapy. Asparagine is widely used in the production of other nutrients and plays an important role in cancer development. Nutritional inhibition therapy targeting asparagine has been used as an anticancer strategy and has shown success in the treatment of leukemia. However, in solid tumors, asparagine restriction alone does not provide ideal therapeutic efficacy. Tumor cells initiate reprogramming processes in response to asparagine deprivation. This review provides a comprehensive overview of asparagine metabolism in cancers. We highlight the physiological role of asparagine and current advances in improving survival and overcoming therapeutic resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
15. FAHD1 and mitochondrial metabolism: a decade of pioneering discoveries.
- Author
-
Cappuccio, Elia, Holzknecht, Max, Petit, Michèle, Heberle, Anne, Rytchenko, Yana, Seretis, Athanasios, Pierri, Ciro L., Gstach, Hubert, Jansen‐Dürr, Pidder, and Weiss, Alexander K. H.
- Subjects
- *
METABOLIC reprogramming , *MITOCHONDRIAL dynamics , *CANCER cell proliferation , *CELLULAR aging , *METABOLIC disorders - Abstract
This review consolidates a decade of research on
fumarylacetoacetate hydrolase domain containing protein 1 (FAHD1), a mitochondrial oxaloacetate tautomerase and decarboxylase with profound implications in cellular metabolism. Despite its critical role as a regulator in mitochondrial metabolism, FAHD1 has remained an often‐overlooked enzyme in broader discussions of mitochondrial function. After more than 12 years of research, it is increasingly clear that FAHD1's contributions to cellular metabolism, oxidative stress regulation, and disease processes such as cancer and aging warrant recognition in both textbooks and comprehensive reviews. The review delves into the broader implications of FAHD1 in mitochondrial function, emphasizing its roles in mitigatingreactive oxygen species (ROS) levels and regulating complex II activity, particularly in cancer cells. This enzyme's significance is further highlighted in the context of aging, where FAHD1's activity has been shown to influence cellular senescence, mitochondrial quality control, and the aging process. Moreover, FAHD1's involvement in glutamine metabolism and its impact on cancer cell proliferation, particularly in aggressive breast cancer subtypes, underscores its potential as a therapeutic target. In addition to providing a comprehensive account of FAHD1's biochemical properties and structural insights, the review integrates emerging hypotheses regarding its role in metabolic reprogramming, immune regulation, and mitochondrial dynamics. By establishing a detailed understanding of FAHD1's physiological roles and therapeutic potential, this work advocates for FAHD1's recognition in foundational texts and resources, marking a pivotal step in its integration into mainstream metabolic research and clinical applications in treating metabolic disorders, cancer, and age‐related diseases. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
16. Metabolism and spatial transcription resolved heterogeneity of glutamine metabolism in cervical carcinoma.
- Author
-
Liu, Qian, Zhu, Jiayu, Abulizi, Guzalinuer, and Hasim, Ayshamgul
- Subjects
- *
DESORPTION electrospray ionization , *MEDICAL sciences , *SQUAMOUS cell carcinoma , *TISSUE metabolism , *CERVICAL cancer - Abstract
Background: Reprogramming of cellular metabolism is a pivotal mechanism employed by tumor cells to facilitate cell growth, proliferation, and differentiation, thereby propelling the progression of cancer. A comprehensive analysis of the transcriptional and metabolic landscape of cervical squamous cell carcinoma (CSCC) at high resolution could greatly enhance the precision of management and therapeutic strategies for this malignancy. Methods: The Air-flow-assisted Desorption Electrospray Ionization Mass Spectro-metric Imaging (AFADESI-MSI) and Spatial Transcriptomics techniques (ST) were employed to investigate the metabolic and transcription profiles of CSCC and normal tissues. For clinical validation, the expression of ASCT2(Ala, Ser, Cys transporter 2) was assessed using immune histochemistry in 122 cases of cervical cancer and 30 cases of cervicitis. Results: The AFADESI-MSI findings have revealed metabolic differences among different CSCC patients. Among them, the metabolic pathways of glutamine show more significant differences. After in situ detection of metabolites, the intensity of glutamate is observed to be significantly higher in cancerous tissue compared to normal tissue, but the intensity is not uniform. To elucidate the potential factors underlying alterations in glutamine metabolism across tissues, we employ ST to quantify mRNA levels. This analysis unveils significant perturbations in glutamine metabolism accompanied by extensive heterogeneity within cervical cancer tissues. After conducting a comprehensive analysis, it has been revealed that the differential expression of ASCT2(encoded by SLC1A5) in distinct regions of cervical cancer tissues plays a pivotal role in inducing heterogeneity in glutamine metabolism. Furthermore, the higher the expression level of ASCT2, the higher the intensity of glutamate is in the region. Further verification, it is found that the expression of ASCT2 protein in CSCC tissues is significantly higher than that in normal tissues (105/122, 86.07%). Conclusions: This finding suggests that the variation in glutamine metabolism is not uniform throughout the tumor. The differential expression of ASCT2 in different regions of cervical cancer tissues seems to play a key role in causing this heterogeneity. This research has opened up new avenues for exploring the glutamine metabolic characteristics of CSCC which is essential for developing more effective targeted therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Macropinocytosis mediates resistance to loss of glutamine transport in triple-negative breast cancer.
- Author
-
Wahi, Kanu, Freidman, Natasha, Wang, Qian, Devadason, Michelle, Quek, Lake-Ee, Pang, Angel, Lloyd, Larissa, Larance, Mark, Zanini, Fabio, Harvey, Kate, O'Toole, Sandra, Guan, Yi Fang, and Holst, Jeff
- Subjects
- *
TRIPLE-negative breast cancer , *CELL metabolism , *PINOCYTOSIS , *GLUTAMINE , *AMINO acids - Abstract
Triple-negative breast cancer (TNBC) metabolism and cell growth uniquely rely on glutamine uptake by the transporter ASCT2. Despite previous data reporting cell growth inhibition after ASCT2 knockdown, we here show that ASCT2 CRISPR knockout is tolerated by TNBC cell lines. Despite the loss of a glutamine transporter and low rate of glutamine uptake, intracellular glutamine steady-state levels were increased in ASCT2 knockout compared to control cells. Proteomics analysis revealed upregulation of macropinocytosis, reduction in glutamine efflux and increased glutamine synthesis in ASCT2 knockout cells. Deletion of ASCT2 in the TNBC cell line HCC1806 induced a strong increase in macropinocytosis across five ASCT2 knockout clones, compared to a modest increase in ASCT2 knockdown. In contrast, ASCT2 knockout impaired cell proliferation in the non-macropinocytic HCC1569 breast cancer cells. These data identify macropinocytosis as a critical secondary glutamine acquisition pathway in TNBC and a novel resistance mechanism to strategies targeting glutamine uptake alone. Despite this adaptation, TNBC cells continue to rely on glutamine metabolism for their growth, providing a rationale for targeting of more downstream glutamine metabolism components. Synopsis: Glutamine uptake fuels triple-negative breast cancer (TNBC) sustaining anabolism and rapid growth, however the underlying mechanisms remain unclear. Here, TNBC cells are shown to tolerate loss of glutamine transporter expression and acquire resistance via inducible acquisition of extracellular carbon sources. Knockout of glutamine transporter ASCT2 leaves human TNBC cell growth unaffected. ASCT2 deletion induces macropinocytosis and utilisation of glucose carbons for glutamine production, restoring intracellular glutamine levels and cell growth. Macropinocytosis-acquired glutamine relies on free amino acids from the culture medium, and is independent of protein breakdown. Macropinocytosis induction is transient and can be rescued by ASCT2 re-expression. Glutamine addiction of malignant mammary epithelial cells relies on compensatory acquisition of extracellular carbon sources. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Microenvironmental G protein‐coupled estrogen receptor‐mediated glutamine metabolic coupling between cancer‐associated fibroblasts and triple‐negative breast cancer cells governs tumour progression.
- Author
-
He, Chongwu, Peng, Meixi, Zeng, Xiaoqiang, Dong, Hanzhi, Sun, Zhengkui, Xu, Jiawei, Liu, Manran, Liu, Liyan, Huang, Yanxiao, Peng, Zhiqiang, Qiu, Yu‐An, Jiang, Chunling, Xu, Bin, and Yu, Tenghua
- Subjects
- *
GLUTAMINE synthetase , *BREAST cancer , *LACTATE dehydrogenase , *TUMOR microenvironment , *ESTROGEN receptors - Abstract
Background: Triple‐negative breast cancer (TNBC) is a particularly aggressive type of breast cancer, known for its lack of effective treatments and unfavorable prognosis. The G protein‐coupled estrogen receptor (GPER), a novel estrogen receptor, is linked to increased malignancy in various cancers. However, its involvement in the metabolic regulation of cancer‐associated fibroblasts (CAFs), a key component in the tumour microenvironment, remains largely unexplored. This study investigates how GPER influences the metabolic interaction between CAFs and TNBC cells, aiming to identify potential therapeutic targets. Methods: The co‐culture system is performed to examine the interaction between CAFs and TNBC cells, with a focus on GPER‐mediated glutamine production and release by CAFs and its subsequent uptake and utilization by TNBC cells. The definite roles of microenvironmental GPER/cAMP/PKA/CREB signalling in regulating the expression of glutamine synthetase (GLUL) and lactate dehydrogenase B (LDHB) are further investigated. Results: Our findings reveal that estrogen‐activated GPER in CAFs significantly upregulates the expression of GLUL and LDHB, leading to increased glutamine production. This glutamine is then secreted into the extracellular matrix and absorbed by TNBC cells, enhancing their viability, motility, and chemoresistance both in vitro and in vivo. TNBC cells further metabolize the glutamine through the glutamine transporter (ASCT2) and glutaminase (GLS1) axes, which, in turn, promote mitochondrial activity and tumour progression. Conclusions: The study identifies GPER as a critical mediator of metabolic coupling between CAFs and TNBC cells, primarily through glutamine metabolism. Targeting the estrogen/GPER/glutamine signalling axis in CAFs offers a promising therapeutic strategy to inhibit TNBC progression and improve patient outcomes. This novel insight into the tumour microenvironment highlights the potential of metabolic interventions in treating TNBC. Key points: Estrogen‐activated GPER in CAFs enhances GLUL and LDHB expression via the cAMP/PKA/CREB signalling, facilitating glutamine production and utilization.Microenvironmental GPER‐induced glutamine serves as a crucial mediator of metabolic coupling between CAFs and TNBC cells, boosting tumour progression by enhancing mitochondrial function.Targeting the glutamine metabolic coupling triggered by estrogen/GPER/GLUL signalling in CAFs is a promising therapeutic strategy for TNBC treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. An immunotherapeutic hydrogel booster inhibits tumor recurrence and promotes wound healing for postoperative management of melanoma
- Author
-
Yuanyuan Yang, Bo Zhang, Yangtao Xu, Wenxiang Zhu, Zinian Zhu, Xibo Zhang, Wenze Wu, Jierong Chen, and Zhiqiang Yu
- Subjects
Glutamine metabolism ,Immunotherapy ,Hydrogel ,Melanoma ,Wound healing ,Materials of engineering and construction. Mechanics of materials ,TA401-492 ,Biology (General) ,QH301-705.5 - Abstract
Low tumor immunogenicity, immunosuppressive tumor microenvironment, and bacterial infections have emerged as significant challenges in postsurgical immunotherapy and skin regeneration for preventing melanoma recurrence. Herein, an immunotherapeutic hydrogel booster (GelMA-CJCNPs) was developed to prevent postoperative tumor recurrence and promote wound healing by incorporating ternary carrier-free nanoparticles (CJCNPs) containing chlorine e6 (Ce6), a BRD4 inhibitor (JQ1), and a glutaminase inhibitor (C968) into methacrylic anhydride-modified gelatin (GelMA) dressings. GelMA-CJCNPs reduced glutathione production by inhibiting glutamine metabolism, thereby preventing the destruction of reactive oxygen species generated by photodynamic therapy, which could amplify oxidative stress to induce severe cell death and enhance immunogenic cell death. In addition, GelMA-CJCNPs reduced M2-type tumor-associated macrophage polarization by blocking glutamine metabolism to reverse the immunosuppressive tumor microenvironment, recruiting more tumor-infiltrating T lymphocytes. GelMA-CJCNPs also downregulated IFN-γ-induced expression of programmed cell death ligand 1 to mitigate acquired immune resistance. Benefiting from the amplified systemic antitumor immunity, GelMA-CJCNPs markedly inhibited the growth of both primary and distant tumors. Moreover, GelMA-CJCNPs demonstrated satisfactory photodynamic antibacterial effects against Staphylococcus aureus infections, thereby promoting postsurgical wound healing. Hence, this immunotherapeutic hydrogel booster, as a facile and effective postoperative adjuvant, possesses a promising potential for inhibiting tumor recurrence and accelerating skin regeneration.
- Published
- 2024
- Full Text
- View/download PDF
20. Glutamine metabolism modulates microglial NLRP3 inflammasome activity through mitophagy in Alzheimer’s disease
- Author
-
Zhixin Zhang, Miao Li, Xiang Li, Zhiyang Feng, Gan Luo, Ying Wang, and Xiaoyan Gao
- Subjects
Alzheimer’s disease ,Microglia ,NLRP3 inflammasome ,Glutamine metabolism ,Mitophagy ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract The NLR family pyrin domain containing 3 (NLRP3) inflammasome in microglia is intimately linked to the pathogenesis of Alzheimer’s disease (AD). Although NLRP3 inflammasome activity is regulated by cellular metabolism, the underlying mechanism remains elusive. Here, we found that under the pathological conditions of AD, the activation of NLRP3 inflammasome in microglia is accompanied by increased glutamine metabolism. Suppression of glutaminase, the rate limiting enzyme in glutamine metabolism, attenuated the NLRP3 inflammasome activation both in the microglia of AD mice and cultured inflammatory microglia. Mechanistically, inhibiting glutaminase blocked the anaplerotic flux of glutamine to the tricarboxylic acid cycle and amino acid synthesis, down-regulated mTORC1 signaling by phosphorylating AMPK, which stimulated mitophagy and limited the accumulation of intracellular reactive oxygen species, ultimately prevented the activation of NLRP3 inflammasomes in activated microglia during AD. Taken together, our findings suggest that glutamine metabolism regulates the activation of NLRP3 inflammasome through mitophagy in microglia, thus providing a potential therapeutic target for AD treatment.
- Published
- 2024
- Full Text
- View/download PDF
21. AMPK Activation in TET2 Downregulated Leukemia Cells Upon Glutamine Limitation
- Author
-
Ahsen Merve BAYRAK and Burcu YUCEL
- Subjects
glutamine metabolism ,tet2 expression ,aml ,ampk ,shrnamediated gene silencing ,Medicine - Abstract
Objective: Metabolic rewiring is a characteristic of cancer cells. Cancer cells require more nutrients for survival and proliferation. Although glutamine can be produced in cells via a series of enzymatic reactions, a group of cancer cells are dependent on extracellular glutamine for survival. TET2 plays a role in DNA demethylation and is a tumor suppressor gene. The TET2 gene is frequently mutated in various cancers, including acute myeloid leukemia (AML). Our study aimed to investigate the association between TET2-knockdown AML cell line HL-60 cells and glutamine metabolism. Methods: To evaluate the association between TET2 expression and glutamine limitation, TET2 was downregulated in HL 60 cells using shRNA plasmids. The proliferation of TET2-knockdown HL-60 cells was calculated in normal and glutamine deficient medium. GLUL mRNA expression was investigated using quantitative reverse transcription polymerase chain reaction and protein levels were evaluated using immunoblotting. Results: The numbers and viability of TET2-knockdown HL-60 cells were decreased in low glutamine-containing medium, but the viability of TET2-knockdown HL-60 cells was higher than that of control cells. GLUL mRNA expressions were increased in TET2- knockdown cells in low glutamine. In addition, P-AMPKα protein expression was increased in TET2-knockdown HL-60 cells in low glutamine-containing medium. Conclusions: Our findings indicate that TET2-knockdown HL-60 cells may be more resistant to glutamine deprivation. In glutamine-deficient medium, the mRNA expression of glutamine synthetase is increased, which could be related to glutamine addiction in cells. In addition, low-glutamyl medium increased the P-AMPKα protein level in TET2-knockdown HL-60 cells.
- Published
- 2024
- Full Text
- View/download PDF
22. KDM4A promotes malignant progression of breast cancer by down-regulating BMP9 inducing consequent enhancement of glutamine metabolism
- Author
-
Yuanxiang Chen, Shiyu Yang, Tao Yu, Tao Zeng, Lan Wei, Yiqing You, Jiafeng Tang, Tingting Dang, Haoli Sun, and Yan Zhang
- Subjects
Breast cancer ,KDM4A ,BMP9 ,Glutamine metabolism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background Recent studies have found that histone-modified genes play an increasingly important role in tumor progression. Lysine(K) specific demethylase 4A (KDM4A) is a histone lysine-specific demethylase highly expressed in a variety of malignant tumors, data showed that KDM4A was negatively correlated with the Bone Morphogenetic Protein 9 (BMP9) in breast cancer. And previous experiments have demonstrated that exogenous BMP9 significantly inhibits breast cancer development. Materials and methods We detected the expression of KDM4A in breast cancer and the relationship between KDM4A and BMP9 using real-time quantitative PCR (RT-qPCR) and Western blot, and verified the interaction between KDM4A and BMP9 by ChIP experiments. At the same time, we also detected whether KDM4A had effects on the RNA and protein stability of BMP9 using actinomycin D and cycloheximide. Measurement of alpha-ketoglutarate (α-KG) level by ELISA to observe the effect of BMP9 on glutamine metabolism in breast cancer cells. Nucleoplasmic distribution of KDM4A after exogenous BMP9 treatment in breast cancer cells were observed by immunofluorescence staining and Western blot. A subcutaneous xenograft tumor model in nude mice was used to study the therapeutic effects of exogenous BMP9 and KDM4A inhibitor (JIB-04) in breast cancer. CCK-8, conoly formation, Transwell, wound healing, and immunohistochemistry were used to monitor the growth of tumor and cell function. Results We found that KDM4A was abnormally highly expressed in breast cancer, and silenced BMP9 expression by removing histone methyl groups from the BMP9 gene region. Meanwhile, KDM4A could also reduce the stability of BMP9 protein. BMP9 inhibit glutamine metabolism in breast cancer, resulting in a decrease in its product α-KG, is confirmed by ELISA. Altered nucleoplasmic distribution of KDM4A due to decreased α-KG was confirmed by immunofluorescence staining and Western blot. Animal experiments confirm that the combination of exogenous BMP9 and JIB-04 shows significantly better results in breast cancer. Conclusions KDM4A silences BMP9 expression by removing histone methyl groups from the BMP9 gene region, leading to further enhancement of glutamine metabolism, which contributes to malignant tumor progression. In addition, using JIB-04 in combination with exogenous BMP9 could inhibit the malignant progression of breast cancer cells and the growth of tumors more significantly.
- Published
- 2024
- Full Text
- View/download PDF
23. 巨噬细胞的谷氨酰胺代谢重编程在 心血管疾病中的作用.
- Author
-
陈幸 and 邓燕
- Abstract
Cardiovascular disease (CVD) poses a significant public health challenge in China, with both incidence and mortality rates increasing. The role of macrophage metabolic reprogramming in CVD has garnered growing attention due to its direct connection to the pathogenesis of the disease and potential therapeutic interventions. Glutamine (GLN), a fundamental energy source for macrophages, plays a crucial role in this context. Alterations in GLN metabolism have substantial effects on macrophage phenotypic transformation, functional performance, and adaptive responses in the context of CVD. Therefore, this review aims to investigate the impact of GLN metabolic reprogramming on macrophages' contribution to the pathogenesis of CVD, specifically focusing on its involvement in atherosclerosis, myocardial infarction, and other cardiovascular conditions. Additionally, it will evaluate the potential of targeting GLN metabolic pathways as a therapeutic strategy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. GLUD1 determines murine muscle stem cell fate by controlling mitochondrial glutamate levels.
- Author
-
Soro-Arnáiz, Inés, Fitzgerald, Gillian, Cherkaoui, Sarah, Zhang, Jing, Gilardoni, Paola, Ghosh, Adhideb, Bar-Nur, Ori, Masschelein, Evi, Maechler, Pierre, Zamboni, Nicola, Poms, Martin, Cremonesi, Alessio, Garcia-Cañaveras, Juan Carlos, De Bock, Katrien, and Morscher, Raphael Johannes
- Subjects
- *
KREBS cycle , *STEM cells , *MUSCLE regeneration , *MUSCLE cells , *METABOLIC regulation - Abstract
Muscle stem cells (MuSCs) enable muscle growth and regeneration after exercise or injury, but how metabolism controls their regenerative potential is poorly understood. We describe that primary metabolic changes can determine murine MuSC fate decisions. We found that glutamine anaplerosis into the tricarboxylic acid (TCA) cycle decreases during MuSC differentiation and coincides with decreased expression of the mitochondrial glutamate deaminase GLUD1. Deletion of Glud1 in proliferating MuSCs resulted in precocious differentiation and fusion, combined with loss of self-renewal in vitro and in vivo. Mechanistically, deleting Glud1 caused mitochondrial glutamate accumulation and inhibited the malate-aspartate shuttle (MAS). The resulting defect in transporting NADH-reducing equivalents into the mitochondria induced compartment-specific NAD+/NADH ratio shifts. MAS activity restoration or directly altering NAD+/NADH ratios normalized myogenesis. In conclusion, GLUD1 prevents deleterious mitochondrial glutamate accumulation and inactivation of the MAS in proliferating MuSCs. It thereby acts as a compartment-specific metabolic brake on MuSC differentiation. [Display omitted] • Glutamine is the main TCA cycle substrate in MuSCs, decreasing with differentiation • Glud1 loss impairs MuSC self-renewal and causes imbalanced differentiation and fusion • Glud1 loss traps mitochondrial glutamate and disrupts the malate-aspartate shuttle (MAS) • Restoring MAS in Glud1- deficient MuSCs blocks early differentiation and fusion imbalance Soro-Arnáiz and Fitzgerald et al. leverage ex vivo and in vivo models to identify how Glud1 determines muscle stem cell (MuSC) fate by controlling mitochondrial glutamate levels and, in turn, the activity of the malate aspartate shuttle. The resulting compartment-specific shift in NAD+/NADH ratios promotes precocious MuSC differentiation and fusion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Activation of the PGC-1α-mediated mitochondrial glutamine metabolism pathway attenuates female offspring osteoarthritis induced by prenatal excessive prednisone.
- Author
-
Li, Qingxian, Zhang, Fan, Dai, Yongguo, Liu, Liang, Chen, Liaobin, and Wang, Hui
- Abstract
Osteoarthritis is a chronic, age-related joint disease. Previous studies have shown that osteoarthritis develops during intrauterine development. Prednisone is frequently used to treat pregnancies complicated by autoimmune diseases. However, limited research has been conducted on the enduring effects of prednisone use during pregnancy on the offspring. In this study, we investigated the effect of excessive prednisone exposure on cartilage development and susceptibility to osteoarthritis in the offspring. We found that prenatal prednisone exposure (PPE) impaired cartilage extracellular matrix (ECM) synthesis, resulting in poor cartilage pathology in female offspring during the adult period, which was further exacerbated after long-distance running stimulation. Additionally, PPE suppressed cartilage development during the intrauterine period. Tracing back to the intrauterine period, we found that Pred, rather than prednisone, decreased glutamine metabolic flux, which resulted in increased oxidative stress, and decreased histone acetylation, and expression of cartilage phenotypic genes. Further, PGC-1α-mediated mitochondrial biogenesis, while PPE caused hypermethylation in the promoter region of PGC-1α and decreased its expression in fetal cartilage by activating the glucocorticoid receptor, resulting in a reduction of glutamine flux controlled by mitochondrial biogenesis. Additionally, overexpression of PGC-1α (either pharmacological or through lentiviral transfection) reversed PPE- and Pred-induced cartilage ECM synthesis impairment. In summary, this study demonstrated that PPE causes chondrodysplasia in female offspring and increases their susceptibility to postnatal osteoarthritis. Hence, targeting PGC-1α early on could be a potential intervention strategy for PPE-induced osteoarthritis susceptibility. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Glutamine metabolism modulates microglial NLRP3 inflammasome activity through mitophagy in Alzheimer's disease.
- Author
-
Zhang, Zhixin, Li, Miao, Li, Xiang, Feng, Zhiyang, Luo, Gan, Wang, Ying, and Gao, Xiaoyan
- Subjects
AMINO acid synthesis ,KREBS cycle ,ALZHEIMER'S disease ,NLRP3 protein ,REACTIVE oxygen species ,GLUTAMINE - Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome in microglia is intimately linked to the pathogenesis of Alzheimer's disease (AD). Although NLRP3 inflammasome activity is regulated by cellular metabolism, the underlying mechanism remains elusive. Here, we found that under the pathological conditions of AD, the activation of NLRP3 inflammasome in microglia is accompanied by increased glutamine metabolism. Suppression of glutaminase, the rate limiting enzyme in glutamine metabolism, attenuated the NLRP3 inflammasome activation both in the microglia of AD mice and cultured inflammatory microglia. Mechanistically, inhibiting glutaminase blocked the anaplerotic flux of glutamine to the tricarboxylic acid cycle and amino acid synthesis, down-regulated mTORC1 signaling by phosphorylating AMPK, which stimulated mitophagy and limited the accumulation of intracellular reactive oxygen species, ultimately prevented the activation of NLRP3 inflammasomes in activated microglia during AD. Taken together, our findings suggest that glutamine metabolism regulates the activation of NLRP3 inflammasome through mitophagy in microglia, thus providing a potential therapeutic target for AD treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Disrupting YAP1-mediated glutamine metabolism induces synthetic lethality alongside ODC1 inhibition in osteosarcoma.
- Author
-
Wang, Hongsheng, Tao, Yining, Han, Jing, Shen, Jiakang, Mu, Haoran, Wang, Zhuoying, Wang, Jinzeng, Jin, Xinmeng, Zhang, Qi, Yang, Yuqin, Lin, Jun, Sun, Mengxiong, Ma, Xiaojun, Ren, Ling, LeBlanc, Amy K., Xu, Jing, Hua, Yingqi, and Sun, Wei
- Subjects
- *
WESTERN immunoblotting , *HIGH throughput screening (Drug development) , *YAP signaling proteins , *REACTIVE oxygen species , *CELL metabolism - Abstract
Purpose: Osteosarcoma, a highly malignant primary bone tumor primarily affecting adolescents, frequently develops resistance to initial chemotherapy, leading to metastasis and limited treatment options. Our study aims to uncover novel therapeutic targets for metastatic and recurrent osteosarcoma. Methods: In this study, we proved the potential of modulating the YAP1-regulated glutamine metabolic pathway to augment the response of OS to DFMO. We initially employed single-cell transcriptomic data to gauge the activation level of polyamine metabolism in MTAP-deleted OS patients. This was further substantiated by transcriptome sequencing data from recurrent and non-recurrent patient tissues, confirming the activation of polyamine metabolism in progressive OS. Through high-throughput drug screening, we pinpointed CIL56, a YAP1 inhibitor, as a promising candidate for a combined therapeutic strategy with DFMO. In vivo, we utilized PDX and CDX models to validate the therapeutic efficacy of this drug combination. In vitro, we conducted western blot analysis, qPCR analysis, immunofluorescence staining, and PuMA experiments to monitor alterations in molecular expression, distribution, and tumor metastasis capability. We employed CCK-8 and colony formation assays to assess the proliferative capacity of cells in the experimental group. We used flow cytometry and reactive oxygen probes to observe changes in ROS and glutamine metabolism within the cells. Finally, we applied RNA-seq in tandem with metabolomics to identify metabolic alterations in OS cells treated with a DFMO and CIL56 combination. This enabled us to intervene and validate the role of the YAP1-mediated glutamine metabolic pathway in DFMO resistance. Results: Through single-cell RNA-seq data analysis, we pinpointed a subset of late-stage OS cells with significantly upregulated polyamine metabolism. This upregulation was further substantiated by transcriptomic profiling of recurrent and non-recurrent OS tissues. High-throughput drug screening revealed a promising combination strategy involving DFMO and CIL56. DFMO treatment curbs the phosphorylation of YAP1 protein in OS cells, promoting nuclear entry and initiating the YAP1-mediated glutamine metabolic pathway. This reduces intracellular ROS levels, countering DFMO's anticancer effect. The therapeutic efficacy of DFMO can be amplified both in vivo and in vitro by combining it with the YAP1 inhibitor CIL56 or the glutaminase inhibitor CB-839. This underscores the significant potential of targeting the YAP1-mediated glutamine metabolic pathway to enhance efficacy of DFMO. Conclusion: Our findings elucidate YAP1-mediated glutamine metabolism as a crucial bypass mechanism against DFMO, following the inhibition of polyamine metabolism. Our study provides valuable insights into the potential role of DFMO in an "One-two Punch" therapy of metastatic and recurrent osteosarcoma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. NEIL3 Upregulated by TFAP2A Promotes M2 Polarization of Macrophages in Liver Cancer via the Mediation of Glutamine Metabolism.
- Author
-
Zhang, Fabiao, Wang, Binfeng, Zhang, Wenlong, Xu, Yongfu, Zhang, Caiming, and Xue, Xiangyang
- Subjects
- *
LIVER cancer , *LIVER cells , *CARRIER proteins , *TUMOR growth , *TUMOR microenvironment - Abstract
Tumor-associated macrophages, which are part of the tumor microenvironment, are a major factor in cancer progression. However, a complete understanding of the regulatory mechanism of M2 polarization of macrophages (Mø) in liver cancer is yet to be established. This study aimed to investigate the potential mechanism by which NEIL3 influenced M2 Mø polarization in liver cancer.Introduction: Bioinformatics analysis analyzed NEIL3 expression and its enriched pathways in liver cancer tissue, as well as its correlation with pathway genes. The upstream transcription factor of NEIL3, TFAP2A, was predicted and its expression in liver cancer tissue was analyzed. The binding relationship between the two was analyzed by dual-luciferase reporter and chromatin immunoprecipitation experiments. qRT-PCR assessed NEIL3 and TFAP2A levels in liver cancer cells. Cell viability was detected by CCK-8, while CD206 and CD86 expression was detected by immunofluorescence. IL-10 and CCR2 expressions were assessed using qRT-PCR, and M2 Mø quantity was detected using flow cytometry. Reagent kits tested glutamine (Gln) consumption, α-ketoglutarate, and glutamate content, as well as NADPH/NADP+ and GSH/GSSG ratios. Expression of Gln transport proteins was detected using Western blot. An animal model was established to investigate the influence of NEIL3 expression on liver cancer growth.Methods: NEIL3 was highly expressed in liver cancer and promoted Mø M2 polarization through Gln metabolism. TFAP2A was identified as the upstream transcription factor of NEIL3 and was highly expressed in liver cancer. Rescue experiments presented that overexpression of NEIL3 reversed the suppressive effect of TFAP2A knockdown on Mø M2 polarization in liver cancer. In vivo experiments demonstrated that the knockdown of NEIL3 could significantly repress the growth of xenograft tumors.Results: This study suggested that the TFAP2A/NEIL3 axis promoted Mø M2 polarization through Gln metabolism, providing a theoretical basis for immune therapy targeting the liver cancer TME. [ABSTRACT FROM AUTHOR]Conclusion: - Published
- 2024
- Full Text
- View/download PDF
29. Circ‐PITX1 promotes non‐small‐cell lung cancer progression through regulating ETS1 expression via miR‐615‐5p.
- Author
-
Guo, Yang, Pan, Jianfang, Gao, Xiaofei, and Zheng, Yan
- Subjects
- *
GLUTAMINE metabolism , *FLOW cytometry , *WOUND healing , *BIOLOGICAL models , *COLONY-forming units assay , *COLORIMETRY , *GLUTAMINE , *T-test (Statistics) , *CIRCULAR RNA , *MICRORNA , *ADENOSINE triphosphate , *CELL proliferation , *TRANSCRIPTION factors , *CELLULAR signal transduction , *REVERSE transcriptase polymerase chain reaction , *XENOGRAFTS , *DESCRIPTIVE statistics , *IN vivo studies , *CANCER patients , *BIOINFORMATICS , *RATS , *IMMUNOHISTOCHEMISTRY , *METABOLISM , *WESTERN immunoblotting , *ONE-way analysis of variance , *ANIMAL experimentation , *LUNG cancer , *DATA analysis software , *DISEASE progression - Abstract
Background: Circular RNAs (circRNAs), produced by reverse splicing, act as important players in human cancers. We aimed to assess the biological functions of circRNA pituitary homeobox 1 (circ‐PITX1) in non‐small‐cell lung cancer (NSCLC). Methods: qRT‐PCR was employed to determine RNA expression. Biological behaviors of NSCLC cells were assessed by CCK‐8, colony formation, EdU assay, flow cytometry, wound healing, and transwell assays. Glutamine catabolism was examined via the measurement of glutamine consumption, α‐ketoglutarate levels, as well as ATP levels. Protein levels were detected by western blot assays. Dual‐luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to reveal the mechanism responsible for circ‐PITX1 regulating NSCLC cell malignancy. The murine xenograft model was established to investigate circ‐PITX1's effect on tumor formation. Results: Circ‐PITX1 was overexpressed in NSCLC tissue samples and cells. Its low expression repressed NSCLC cell proliferation and motility. Moreover, our data revealed its downregulation inhibited glutamine catabolism and tumor formation and promoted cell apoptosis. In addition, circ‐PITX1 bound to miR‐615‐5p, and its inhibitory effect on tumor cellular behaviors could be reversed after decreasing miR‐615‐5p expression. The miRNA targeted E26 transformation specific‐1 (ETS1), whose upregulation abolished miR‐615‐5p overexpression‐induced effects in NSCLC cells. Furthermore, circ‐PITX1 positively modulated ETS1 production through interaction with miR‐615‐5p. Conclusion: Circ‐PITX1 facilitated NSCLC progression via modulating miR‐615‐5p/ETS1 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. KDM4A promotes malignant progression of breast cancer by down-regulating BMP9 inducing consequent enhancement of glutamine metabolism.
- Author
-
Chen, Yuanxiang, Yang, Shiyu, Yu, Tao, Zeng, Tao, Wei, Lan, You, Yiqing, Tang, Jiafeng, Dang, Tingting, Sun, Haoli, and Zhang, Yan
- Subjects
CANCER cell growth ,BONE morphogenetic proteins ,BREAST cancer ,PROTEIN stability ,GENE expression ,GLUTAMINE - Abstract
Background: Recent studies have found that histone-modified genes play an increasingly important role in tumor progression. Lysine(K) specific demethylase 4A (KDM4A) is a histone lysine-specific demethylase highly expressed in a variety of malignant tumors, data showed that KDM4A was negatively correlated with the Bone Morphogenetic Protein 9 (BMP9) in breast cancer. And previous experiments have demonstrated that exogenous BMP9 significantly inhibits breast cancer development. Materials and methods: We detected the expression of KDM4A in breast cancer and the relationship between KDM4A and BMP9 using real-time quantitative PCR (RT-qPCR) and Western blot, and verified the interaction between KDM4A and BMP9 by ChIP experiments. At the same time, we also detected whether KDM4A had effects on the RNA and protein stability of BMP9 using actinomycin D and cycloheximide. Measurement of alpha-ketoglutarate (α-KG) level by ELISA to observe the effect of BMP9 on glutamine metabolism in breast cancer cells. Nucleoplasmic distribution of KDM4A after exogenous BMP9 treatment in breast cancer cells were observed by immunofluorescence staining and Western blot. A subcutaneous xenograft tumor model in nude mice was used to study the therapeutic effects of exogenous BMP9 and KDM4A inhibitor (JIB-04) in breast cancer. CCK-8, conoly formation, Transwell, wound healing, and immunohistochemistry were used to monitor the growth of tumor and cell function. Results: We found that KDM4A was abnormally highly expressed in breast cancer, and silenced BMP9 expression by removing histone methyl groups from the BMP9 gene region. Meanwhile, KDM4A could also reduce the stability of BMP9 protein. BMP9 inhibit glutamine metabolism in breast cancer, resulting in a decrease in its product α-KG, is confirmed by ELISA. Altered nucleoplasmic distribution of KDM4A due to decreased α-KG was confirmed by immunofluorescence staining and Western blot. Animal experiments confirm that the combination of exogenous BMP9 and JIB-04 shows significantly better results in breast cancer. Conclusions: KDM4A silences BMP9 expression by removing histone methyl groups from the BMP9 gene region, leading to further enhancement of glutamine metabolism, which contributes to malignant tumor progression. In addition, using JIB-04 in combination with exogenous BMP9 could inhibit the malignant progression of breast cancer cells and the growth of tumors more significantly. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. ASCT2 Regulates Fatty Acid Metabolism to Trigger Glutamine Addiction in Basal-like Breast Cancer.
- Author
-
Wang, Jia, Zhang, Qian, Fu, Huaizi, Han, Yi, Li, Xue, Zou, Qianlin, Yuan, Shengtao, and Sun, Li
- Subjects
- *
GLUTAMINE metabolism , *LIPID metabolism , *EARLY medical intervention , *CARRIER proteins , *RESEARCH funding , *BREAST tumors , *REVERSE transcriptase polymerase chain reaction , *DESCRIPTIVE statistics , *MICE , *GENE expression , *ANIMAL experimentation , *WESTERN immunoblotting , *FATTY acids , *PEROXISOME proliferator-activated receptors ,BREAST tumor prevention - Abstract
Simple Summary: In recent years, cancer has gradually become a terrible killer of human health. Due to various factors such as tumor heterogeneity and the complexity of the microenvironment, completely conquering cancer has always been a major problem in the scientific research community. Therefore, more detailed research and individualized treatment are essential. Everyone knows that tumor cells in the process of rapid proliferation need replenishing nutrients to sustain their energy supply. Intervention of tumor cell metabolism is gradually becoming a way to treat cancer. As an important transporter of glutamine, ASCT2 can uptake glutamine from the tumor microenvironment to support the growth and metabolism of cancer cells. Deficiency of ASCT2 can significantly inhibit tumor progression. However, not all patients benefit from this treatment. Our study aims to explore the metabolic heterogeneity of breast cancer and its related mechanisms, as well as to find metabolic-sensitive populations for further precision treatment. As a crucial amino acid, glutamine can provide the nitrogen and carbon sources needed to support cancer cell proliferation, invasion, and metastasis. Interestingly, different types of breast cancer have different dependences on glutamine. This research shows that basal-like breast cancer depends on glutamine, while the other types of breast cancer may be more dependent on glucose. Glutamine transporter ASCT2 is highly expressed in various cancers and significantly promotes the growth of breast cancer. However, the key regulatory mechanism of ASCT2 in promoting basal-like breast cancer progression remains unclear. Our research demonstrates the significant change in fatty acid levels caused by ASCT2, which may be a key factor in glutamine sensitivity. This phenomenon results from the mutual activation between ASCT2-mediated glutamine transport and lipid metabolism via the nuclear receptor PPARα. ASCT2 cooperatively promoted PPARα expression, leading to the upregulation of lipid metabolism. Moreover, we also found that C118P could inhibit lipid metabolism by targeting ASCT2. More importantly, this research identifies a potential avenue of evidence for the prevention and early intervention of basal-like breast cancer by blocking the glutamine–lipid feedback loop. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. HJURP Derived from Cancer-Associated Fibroblasts Promotes Glutamine Metabolism to Induce Resistance to Doxorubicin in Ovarian Cancer.
- Author
-
Yanfang Lan, Hao Xu, and Lanying Jin
- Abstract
Cancer-associated fibroblasts (CAFs) are closely associated with tumor drug resistance. This study intended to delineate how CAFs induced DOX resistance in ovarian cancer. Differential gene expression analysis of ovarian cancer CAFs was completed using Gene Expression Omnibus database. CAFs and normal fibroblasts (NFs) were isolated from ovarian cancer tissues and adjacent normal tissues. The expressions of Holliday Junction Recognition Protein (HJURP), α -smooth muscle actin (α -SMA), and fibroblast activation protein alpha (FAP) were assessed by quantitative reverse transcription polymerase chain reaction and Western blot (WB), α -SMA and FAP were detected by immunofluorescence. A2780 cells were treated with CAF or NF conditioned medium (CM), and protein expression of HJURP was assessed by WB. A2780-DOX cells were constructed and cultured with CAF or NF CM, and cell viability and IC50 value of DOX were assayed by Cell Counting Kit-8. Kits were used to test glutamine metabolism and mitochondrial tricarboxylic acid (TCA) cycle products, while WB was utilized to assess expressions of amino acid transporters. mRNA and protein levels of HJURP in CAFs derived from ovarian cancer were significantly higher than those in NFs. Culturing ovarian cancer cells with CAF CM could increase protein expressions of HJURP. HJURP derived from CAFs significantly enhanced viability of A2780-DOX cells and DOX resistance. CAF-derived HJURP fostered glutamine metabolism and mitochondrial TCA cycle in ovarian cancer resistant cells ultimately leading to ovarian cancer DOX resistance. CAF-derived HJURP drove ovarian cancer glutamine metabolism and DOX resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Glutamine and leukemia research: progress and clinical prospects.
- Author
-
Wang, Zexin, Liu, Miao, and Yang, Qiang
- Subjects
LEUCOCYTES ,LYMPHOBLASTIC leukemia ,ACUTE myeloid leukemia ,ACUTE leukemia ,APOPTOSIS inhibition - Abstract
Leukemia is an abnormal proliferation of white blood cells that occurs in bone marrow and expands through the blood. It arises from dysregulated differentiation, uncontrolled growth, and inhibition of apoptosis. Glutamine (GLN) is a "conditionally essential" amino acid that promotes growth and proliferation of leukemic cells. Recently, details about the role of GLN and its metabolism in the diagnosis and treatment of acute myeloid, chronic lymphocytic, and acute lymphoblastic leukemia have emerged. The uptake of GLN by leukemia cells and the dynamic changes of glutamine-related indexes in leukemia patients may be able to assist in determining whether the condition of leukemia is in a state of progression, remission or relapse. Utilizing the possible differences in GLN metabolism in different subtypes of leukemia may help to differentiate between different subtypes of leukemia, thus providing a basis for accurate diagnosis. Targeting GLN metabolism in leukemia requires simultaneous blockade of multiple metabolic pathways without interfering with the normal cellular and immune functions of the body to achieve effective leukemia therapy. The present review summarizes recent advances, possible applications, and clinical perspectives of GLN metabolism in leukemia. In particular, it focuses on the prospects of GLN metabolism in the diagnosis and treatment of acute myeloid leukemia. The review provides new directions and hints at potential roles for future clinical treatments and studies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. AMPK Activation in TET2 Downregulated Leukemia Cells Upon Glutamine Limitation.
- Author
-
BAYRAK, Ahsen Merve and YUCEL, Burcu
- Subjects
REVERSE transcriptase polymerase chain reaction ,TUMOR suppressor genes ,DNA demethylation ,ACUTE myeloid leukemia ,GENE expression - Abstract
Copyright of Medeniyet Medical Journal is the property of Galenos Yayinevi Tic. LTD. STI and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
35. Emerging roles of the chromatin remodeler MORC2 in cancer metabolism.
- Author
-
Mohapatra, Bibhukalyan and Pakala, Suresh B.
- Abstract
Cancer is characterized by metabolic reprogramming in cancer cells, which is crucial for tumorigenesis. The highly deregulated chromatin remodeler MORC2 contributes to cell proliferation, invasion, migration, DNA repair, and chemoresistance. MORC2 also plays a key role in metabolic reprogramming, including lipogenesis, glucose, and glutamine metabolism. A recent study showed that MORC2-regulated glucose metabolism affects the expression of E-cadherin, a crucial protein in the epithelial-to-mesenchymal transition. This review discusses recent developments in MORC2 regulated cancer cell metabolism and its role in cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Inhibition of Glutamate‐to‐Glutathione Flux Promotes Tumor Antigen Presentation in Colorectal Cancer Cells
- Author
-
Tao Yu, Kevin Van der Jeught, Haiqi Zhu, Zhuolong Zhou, Samantha Sharma, Sheng Liu, Haniyeh Eyvani, Ka Man So, Naresh Singh, Jia Wang, George E. Sandusky, Yunlong Liu, Mateusz Opyrchal, Sha Cao, Jun Wan, Chi Zhang, and Xinna Zhang
- Subjects
colorectal cancer ,glutamine metabolism ,immune checkpoint blockade ,immunoproteasome ,MHC‐I antigen presentation ,single‐cell flux estimation analysis ,Science - Abstract
Abstract Colorectal cancer (CRC) cells display remarkable adaptability, orchestrating metabolic changes that confer growth advantages, pro‐tumor microenvironment, and therapeutic resistance. One such metabolic change occurs in glutamine metabolism. Colorectal tumors with high glutaminase (GLS) expression exhibited reduced T cell infiltration and cytotoxicity, leading to poor clinical outcomes. However, depletion of GLS in CRC cells has minimal effect on tumor growth in immunocompromised mice. By contrast, remarkable inhibition of tumor growth is observed in immunocompetent mice when GLS is knocked down. It is found that GLS knockdown in CRC cells enhanced the cytotoxicity of tumor‐specific T cells. Furthermore, the single‐cell flux estimation analysis (scFEA) of glutamine metabolism revealed that glutamate‐to‐glutathione (Glu‐GSH) flux, downstream of GLS, rather than Glu‐to‐2‐oxoglutarate flux plays a key role in regulating the immune response of CRC cells in the tumor. Mechanistically, inhibition of the Glu‐GSH flux activated reactive oxygen species (ROS)‐related signaling pathways in tumor cells, thereby increasing the tumor immunogenicity by promoting the activity of the immunoproteasome. The combinatorial therapy of Glu‐GSH flux inhibitor and anti‐PD‐1 antibody exhibited a superior tumor growth inhibitory effect compared to either monotherapy. Taken together, the study provides the first evidence pointing to Glu‐GSH flux as a potential therapeutic target for CRC immunotherapy.
- Published
- 2025
- Full Text
- View/download PDF
37. Rotavirus rewires host cell metabolic pathways toward glutamine catabolism for effective virus infection
- Author
-
Suvrotoa Mitra, Ratul Datta Chaudhuri, Rakesh Sarkar, Shreya Banerjee, Arpita Mukherjee, Ranjana Sharma, Animesh Gope, Kei Kitahara, Shin-Ichi Miyoshi, and Mamta Chawla-Sarkar
- Subjects
Rotavirus ,metabolomics ,glutamine metabolism ,aspartate aminotransferase ,antiviral ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Rotavirus (RV) accounts for 19.11% of global diarrheal deaths. Though GAVI assisted vaccine introduction has curtailed RV induced mortality, factors like RV strain diversity, differential infantile gut microbiome, malnutrition, interference from maternal antibodies and other administered vaccines, etc. often compromise vaccine efficacy. Herein emerges the need of antivirals which can be administered adjunct to vaccination to curb the socio-economic burden stemming from frequent RV infection. Cognisance of pathogen-perturbed host cellular physiology has revolutionized translational research and aided precision-based therapy, particularly for viruses, with no metabolic machinery of their own. To date there has been limited exploration of the host cellular metabolome in context of RV infection. In this study, we explored the endometabolomic landscape of human intestinal epithelial cells (HT-29) on RV-SA11 infection. Significant alteration of host cellular metabolic pathways like the nucleotide biosynthesis pathway, alanine, aspartate and glutamate metabolism pathway, the host citric acid cycle was observed in RV-SA11 infection scenario. Detailed study further revealed that RV replication is exclusively dependent on glutamine metabolism for their propagation in host cells. Glutamine metabolism generates glutamate, aspartate, and asparagine which facilitates virus infection. Abrogation of aspartate biogenesis from glutamine by use of Aminooxyacetic acid (AOAA), significantly curbed RV-SA11 infection in-vitro and in-vivo. Overall, the study improves our understanding of host-rotavirus interactome and recognizes host glutamine metabolism pathway as a suitable target for effective therapeutic intervention against RV infection.
- Published
- 2024
- Full Text
- View/download PDF
38. Glutamine and leukemia research: progress and clinical prospects
- Author
-
Zexin Wang, Miao Liu, and Qiang Yang
- Subjects
Glutamine ,Leukemia ,Glutamine metabolism ,Glutaminase ,Glutamine transporter protein ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Leukemia is an abnormal proliferation of white blood cells that occurs in bone marrow and expands through the blood. It arises from dysregulated differentiation, uncontrolled growth, and inhibition of apoptosis. Glutamine (GLN) is a "conditionally essential" amino acid that promotes growth and proliferation of leukemic cells. Recently, details about the role of GLN and its metabolism in the diagnosis and treatment of acute myeloid, chronic lymphocytic, and acute lymphoblastic leukemia have emerged. The uptake of GLN by leukemia cells and the dynamic changes of glutamine-related indexes in leukemia patients may be able to assist in determining whether the condition of leukemia is in a state of progression, remission or relapse. Utilizing the possible differences in GLN metabolism in different subtypes of leukemia may help to differentiate between different subtypes of leukemia, thus providing a basis for accurate diagnosis. Targeting GLN metabolism in leukemia requires simultaneous blockade of multiple metabolic pathways without interfering with the normal cellular and immune functions of the body to achieve effective leukemia therapy. The present review summarizes recent advances, possible applications, and clinical perspectives of GLN metabolism in leukemia. In particular, it focuses on the prospects of GLN metabolism in the diagnosis and treatment of acute myeloid leukemia. The review provides new directions and hints at potential roles for future clinical treatments and studies.
- Published
- 2024
- Full Text
- View/download PDF
39. Glutamine Metabolism and Prostate Cancer.
- Author
-
Erb, Holger H. H., Polishchuk, Nikita, Stasyk, Oleh, Kahya, Uğur, Weigel, Matthias M., and Dubrovska, Anna
- Subjects
- *
GLUTAMINE metabolism , *GLUTAMINE , *PATIENT safety , *CLINICAL trials , *PROSTATE tumors , *CANCER chemotherapy , *METABOLISM , *ONCOGENES , *DRUG efficacy , *DISEASE progression , *ANDROGEN receptors , *DRUG resistance , *CHEMICAL inhibitors - Abstract
Simple Summary: Glutamine (Gln) plays a critical role in the development and progression of prostate cancer (PCa). Non-malignant prostate cells are not addicted to Gln, but PCa cells become Gln-dependent due to the low levels of the Gln-producing enzyme glutamine synthetase. Gln metabolism in PCa is controlled by oncogenes such as MYC, AR, and mTOR, which contribute to therapy resistance and more aggressive forms of the disease. Studies have shown that depriving PCa cells of Gln can inhibit their growth and survival and make them more sensitive to radiotherapy. Targeting Gln metabolism is, therefore, a promising approach for PCa treatment. Clinical trials are currently testing the safety and efficacy of drugs that inhibit Gln metabolism in combination with other therapies for different types of cancer, including PCa. Understanding of how tumor cells metabolically interact with their environment may help improve these treatments and patient outcomes. In this review, we present the latest advances in Gln research in PCa and insights into the current clinical trials. Glutamine (Gln) is a non-essential amino acid that is involved in the development and progression of several malignancies, including prostate cancer (PCa). While Gln is non-essential for non-malignant prostate epithelial cells, PCa cells become highly dependent on an exogenous source of Gln. The Gln metabolism in PCa is tightly controlled by well-described oncogenes such as MYC, AR, and mTOR. These oncogenes contribute to therapy resistance and progression to the aggressive castration-resistant PCa. Inhibition of Gln catabolism impedes PCa growth, survival, and tumor-initiating potential while sensitizing the cells to radiotherapy. Therefore, given its significant role in tumor growth, targeting Gln metabolism is a promising approach for developing new therapeutic strategies. Ongoing clinical trials evaluate the safety and efficacy of Gln catabolism inhibitors in combination with conventional and targeted therapies in patients with various solid tumors, including PCa. Further understanding of how PCa cells metabolically interact with their microenvironment will facilitate the clinical translation of Gln inhibitors and help improve therapeutic outcomes. This review focuses on the role of Gln in PCa progression and therapy resistance and provides insights into current clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. Radiation‐induced exosomes promote oral squamous cell carcinoma progression via enhancing SLC1A5‐glutamine metabolism.
- Author
-
Yang, Rongchun, Zhang, Siyuan, Wang, Lixuan, Chen, Yingyao, Chen, Xiaobing, Xia, Juan, Ren, Xianyue, Cheng, Bin, and Chen, Xijuan
- Subjects
- *
CANCER cell proliferation , *SQUAMOUS cell carcinoma , *EXTRACELLULAR vesicles , *CELLULAR aging , *CANCER cells - Abstract
Background: Radiotherapy (RT) can drive cancer cells to enter a state of cellular senescence in which cells can secrete senescence‐associated secretory phenotype (SASP) and produce small extracellular vesicles (sEVs) to interact with cells in the tumor microenvironment (TME). Tumor‐derived sEVs that are taken up by recipient cells contribute to cancer cell metabolic plasticity, resistance to anticancer therapy, and adaptation to the TME. However, how radiation‐induced sEVs support oral squamous cell carcinoma (OSCC) progression remains unclear. Methods: Beta‐galactosidase staining and SASP mRNA expression analysis were used to evaluate the senescence‐associated activity of OSCC cells after irradiation. Nanoparticle tracking analysis was performed to identify radiation‐induced sEVs. Liquid chromatography–tandem mass spectrometry (LC–MS) was used to explore changes in the levels of proteins in radiation‐induced sEVs. Cell Counting Kit‐8 and colony formation assays were performed to investigate the function of radiation‐induced SASP and sEVs in vitro. A xenograft tumor model was established to investigate the functions of radiation‐induced sEVs and V‐9302 in vivo as well as the underlying mechanisms. Bioinformatics analysis was performed to determine the relationship between glutamine metabolism and OSCC recurrence. Results: We determined that the radiation‐induced SASP triggered OSCC cell proliferation. Additionally, radiation‐induced sEVs exacerbated OSCC cell malignancy. LC–MS/MS and bioinformatics analyses revealed that SLC1A5, which is a cellular receptor that participates in glutamine uptake, was significantly enriched in radiation‐induced sEVs. In vitro and in vivo, inhibiting SLC1A5 could block the oncogenic effects of radiation‐induced sEVs in OSCC. Conclusion: Radiation‐induced sEVs might promote the proliferation of unirradiated cancer cells by enhancing glutamine metabolism; this might be a novel molecular mechanism underlying radiation resistance in OSCC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. TFAP2A Activates S100A2 to Mediate Glutamine Metabolism and Promote Lung Adenocarcinoma Metastasis.
- Author
-
Zeng, Tao, Ren, Wangsheng, Zeng, Hang, Wang, Dachun, Wu, Xianyu, and Xu, Guo
- Subjects
- *
CELL metabolism , *CELL migration , *CELL survival , *METABOLIC disorders , *METASTASIS , *GLUTAMINE - Abstract
Background: Lung adenocarcinoma (LUAD) is a fatal disease with metabolic abnormalities. The dysregulation of S100 calcium‐binding protein A2 (S100A2), a member of the S100 protein family, is connected to the development of various cancers. The impact of S100A2 on the LUAD occurrence and metastasis, however, has not yet been reported. The functional mechanism of S100A2 on LUAD cell metastasis was examined in this article. Methods: The expression of TFAP2A and S100A2 in LUAD tissues and cells was analyzed by bioinformatics and qRT‐PCR, respectively. The enrichment pathway analysis was performed on S100A2. Bioinformatics analysis determined the binding relationship between TFAP2A and S100A2, and their interaction was validated through dual‐luciferase and chromatin immunoprecipitation experiments. Cell viability was determined using cell counting kit‐8 (CCK‐8). A transwell assay was performed to analyze the invasion and migration of cells. Immunofluorescence was conducted to obtain vimentin and E‐cadherin expression, and a western blot was used to detect the expression of MMP‐2, MMP‐9, GLS, and GLUD1. The kits measured the NADPH/NADP ratio, glutathione (GSH)/glutathione disulfide (GSSG) levels, and the contents of glutamine, α‐KG, and glutamate. Results: S100A2 was upregulated in LUAD tissues and cells, and S100A2 mediated glutamine metabolism to induce LUAD metastasis. Additionally, the transcriptional regulator TFAP2A was discovered upstream of S100A2, and TFAP2A expression was upregulated in LUAD, which indicated that TFAP2A promoted the S100A2 expression. The rescue experiment found that upregulation of S100A2 could reverse the inhibitory effects of silencing TFAP2A on glutamine metabolism and cell metastasis. Conclusion: In conclusion, by regulating glutamine metabolism, the TFAP2A/S100A2 axis facilitated LUAD metastasis. This suggested that targeting S100A2 could be beneficial for LUAD treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Pyruvate Carboxylase Attenuates Myocardial Ischemia–Reperfusion Injury in Heart Transplantation via Wnt/β-Catenin-Mediated Glutamine Metabolism.
- Author
-
Wang, Zihao, Lan, Hongwen, Wang, Yixuan, Zheng, Qiang, Li, Chenghao, Wang, Kan, Xiong, Tixiusi, Wu, Qingping, and Dong, Nianguo
- Subjects
HEART transplantation ,PYRUVATE carboxylase ,BAX protein ,HEART diseases ,BCL-2 proteins - Abstract
The ischemia–reperfusion process of a donor heart during heart transplantation leads to severe mitochondrial dysfunction, which may be the main cause of donor heart dysfunction after heart transplantation. Pyruvate carboxylase (PC), an enzyme found in mitochondria, is said to play a role in the control of oxidative stress and the function of mitochondria. This research examined the function of PC and discovered the signaling pathways controlled by PC in myocardial IRI. We induced IRI using a murine heterotopic heart transplantation model in vivo and a hypoxia–reoxygenation cell model in vitro and evaluated inflammatory responses, oxidative stress levels, mitochondrial function, and cardiomyocyte apoptosis. In both in vivo and in vitro settings, we observed a significant decrease in PC expression during myocardial IRI. PC knockdown aggravated IRI by increasing MDA content, LDH activity, TUNEL-positive cells, serum cTnI level, Bax protein expression, and the level of inflammatory cytokines and decreasing SOD activity, GPX activity, and Bcl-2 protein expression. PC overexpression yielded the opposite findings. Additional research indicated that reducing PC levels could block the Wnt/β-catenin pathway and glutamine metabolism by hindering the movement of β-catenin to the nucleus and reducing the activity of complex I and complex II, as well as ATP levels, while elevating the ratios of NADP+/NADPH and GSSG/GSH. Overall, the findings indicated that PC therapy can shield the heart from IRI during heart transplantation by regulating glutamine metabolism through the Wnt/β-catenin pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Deciphering glutamine metabolism patterns for malignancy and tumor microenvironment in clear cell renal cell carcinoma.
- Author
-
Wu, Gengrun, Li, Teng, Chen, Yuanbiao, Ye, Shiqi, Zhou, Siqi, Tian, Xi, Anwaier, Aihetaimujiang, Zhu, Shuxuan, Xu, Wenhao, Hao, Xiaohang, Ye, Dingwei, and Zhang, Hailiang
- Subjects
- *
RENAL cell carcinoma , *TUMOR microenvironment , *GLUTAMINE , *METABOLIC reprogramming , *METABOLISM - Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer characterized by metabolic reprogramming. Glutamine metabolism is pivotal in metabolic reprogramming, contributing to the significant heterogeneity observed in ccRCC. Consequently, developing prognostic markers associated with glutamine metabolism could enhance personalized treatment strategies for ccRCC patients. This study obtained RNA sequencing and clinical data from 763 ccRCC cases sourced from multiple databases. Consensus clustering of 74 glutamine metabolism related genes (GMRGs)- profiles stratified the patients into three clusters, each of which exhibited distinct prognosis, tumor microenvironment, and biological characteristics. Then, six genes (SMTNL2, MIOX, TMEM27, SLC16A12, HRH2, and SAA1) were identified by machine-learning algorithms to develop a predictive signature related to glutamine metabolism, termed as GMRScore. The GMRScore showed significant differences in clinical prognosis, expression profile of immune checkpoints, abundance of immune cells, and immunotherapy response of ccRCC patients. Besides, the nomogram incorporating the GMRScore and clinical features showed strong predictive performance in prognosis of ccRCC patients. ALDH18A1, one of the GRMGs, exhibited elevated expression level in ccRCC and was related to markedly poorer prognosis in the integrated cohort, validated by proteomic profiling of 232 ccRCC samples from Fudan University Shanghai Cancer Center (FUSCC). Conducting western blotting, CCK-8, transwell, and flow cytometry assays, we found the knockdown of ALDH18A1 in ccRCC significantly promoted apoptosis and inhibited proliferation, invasion, and epithelial-mesenchymal transition (EMT) in two human ccRCC cell lines (786-O and 769-P). In conclusion, we developed a glutamine metabolism-related prognostic signature in ccRCC, which is tightly linked to the tumor immune microenvironment and immunotherapy response, potentially facilitating precision therapy for ccRCC patients. Additionally, this study revealed the key role of ALDH18A1 in promoting ccRCC progression for the first time. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. 肝纤维化进展中脾脏对肝脏巨噬细胞 活化肝星状细胞作用的影响.
- Author
-
张少颖, 万丹, 邓熙, 梁肖, 梁凡凡, 张冲宇, 朱佳真, 赵阳, and 李宗芳
- Abstract
Objective To investigate the effect of spleen on hepatic macrophages mediated activation of hepatic stellate cells(HSCs)in mice with liver fibrosis. Methods Eighteen male C57BL/6 mice were randomly divided into three groups. Mice in Group A and Group B were injected intraperitoneally with CCl4 to establish liver fibrosis mouse model, while those in Group C were injected with corn oil as normal control. Four weeks later, mice with liver fibrosis received splenectomy(Spx)or sham operation(Sham), respectively. After continuous injection for 2 weeks, liver homogenates(L-Homo)were prepared and liver cells were isolated from the three groups. Expressions of IL-1β, IL-13, TGF-β, TNF-α, PDGF-β and VEGF in the liver homogenates of the three groups were detected by Luminex multifactor analysis. The expressions of these cytokines in liver macrophages(L-Mφ)and other non-parenchymal cells of Sham and Spx mice were analyzed by Real-time quantitative PCR(RT-qPCR)and flow cytometry. Macrophage cell line RAW264.7 or bone marrow-derived macrophages(BMDMs)were treated with liver homogenates from the Sham and Spx groups. Then the differently treated RAW264.7 cells were analyzed for mRNA expressions of cytokines and glutamine metabolism-related molecules by RT-qPCR, or transwell co-cultured with hepatic stellate cell line JS1. After co-culture, the survival and extracellular matrix expression of JS1 cells were analyzed. For comparison, Student's t test(between two groups)or one-way analysis of variance(among multiple groups)were used. Results Compared with normal control group, the concentrations of IL-1β, IL-13, TGF-β and TNF-α in the L-Homo of model group were significantly increased and showed higher levels in Sham group than in Spx group. Moreover, the hepatic macrophages were indicated as the major source of these cytokines. Consistently, macrophages treated with liver homogenate of Sham mice had increased expressions of IL-1β, TGF-β and TNF-α and glutaminase(GLS). After co-culture with macrophages treated with liver homogenate of Sham group rather than Spx group, JS1 expressed higher expressions of α-SMA and collagens. Conclusion The spleen is involved in regulating the secretion of cytokines by hepatic macrophages and enhancing their ability to activate hepatic stellate cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. MRPL35 Induces Proliferation, Invasion, and Glutamine Metabolism in NSCLC Cells by Upregulating SLC7A5 Expression.
- Author
-
Hou, Wei, Chen, Juan, and Wang, Yaoyuan
- Subjects
- *
GLUTAMINE , *CELL metabolism , *NON-small-cell lung carcinoma , *DEUBIQUITINATING enzymes - Abstract
Background: Mitochondrial ribosomal protein L35 (MRPL35) has been reported to contribute to the growth of non–small cell lung cancer (NSCLC) cells. However, the functions and mechanisms of MRPL35 on glutamine metabolism in NSCLC remain unclear. Methods: The detection of mRNA and protein of MRPL35, ubiquitin‐specific protease 39 (USP39), and solute carrier family 7 member 5 (SLC7A5) was conducted using qRT‐PCR and western blotting. Cell proliferation, apoptosis, and invasion were evaluated using the MTT assay, EdU assay, flow cytometry, and transwell assay, respectively. Glutamine metabolism was analyzed by detecting glutamine consumption, α‐ketoglutarate level, and glutamate production. Cellular ubiquitination analyzed the deubiquitination effect of USP39 on MRPL35. An animal experiment was conducted for in vivo analysis. Results: MRPL35 was highly expressed in NSCLC tissues and cell lines, and high MRPL35 expression predicted poor outcome in NSCLC patients. In vitro analyses suggested that MRPL35 knockdown suppressed NSCLC cell proliferation, invasion, and glutamine metabolism. Moreover, MRPL35 silencing hindered tumor growth in vivo. Mechanistically, USP39 stabilized MRPL35 expression by deubiquitination and then promoted NSCLC cell proliferation, invasion, and glutamine metabolism. In addition, MRPL35 positively affected SLC7A5 expression in NSCLC cells in vitro and in vivo. Moreover, the anticancer effects of MRPL35 silencing could be rescued by SLC7A5 overexpression in NSCLC cells. Conclusion: MRPL35 expression was stabilized by USP39‐induced deubiquitination in NSCLC cells, and knockdown of MRPL35 suppressed NSCLC cell proliferation, invasion, and glutamine metabolism in vitro and impeded tumor growth in vivo by upregulating SLC7A5, providing a promising therapeutic target for NSCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Cancer-associated fibroblasts-derived exosomal METTL3 promotes the proliferation, invasion, stemness and glutaminolysis in non-small cell lung cancer cells by eliciting SLC7A5 m6A modification.
- Author
-
Fan, Yafeng and Yu, Yanling
- Subjects
NON-small-cell lung carcinoma ,EXOSOMES ,CANCER cells ,WESTERN immunoblotting ,GENE expression - Abstract
Cancer-associated fibroblasts (CAFs) can promote the crosstalk between cancer cells and tumor microenvironment by exosomes. METTL3-mediated N6-methyladenine (m6A) modification has been proved to promote the progression of non-small cell lung cancer (NSCLC). Here, we focused on the impacts of CAFs-derived exosomes and METTL3-mediated m6A modification on NSCLC progression. Functional analyses were conducted using Cell Counting Kit-8, EdU, colony formation, sphere formation and transwell assays, respectively. Glutamine metabolism was evaluated by detecting glutamate consumption, and the production of intercellular glutamate and α-ketoglutarate (α-KG). qRT-PCR and western blotting analyses were utilized to measure the levels of genes and proteins. Exosomes were isolated by kits. The methylated RNA immunoprecipitation assay detected the m6A modification profile of Amino acid transporter LAT1 (SLC7A5) mRNA. The NSCLC mouse model was established to conduct in vivo experiments. We found that CAFs promoted the proliferation, invasion, stemness and glutaminolysis in NSCLC cells. METTL3 was enriched in CAFs and was packaged into exosomes. After knockdown of METTL3 in CAF exosomes, it was found the oncogenic effects of CAFs on NSCLC cells were suppressed. CAFs elevated m6A levels in NSCLC cells. Mechanistically, exosomal METTL3-induced m6A modification in SLC7A5 mRNA and stabilized its expression in NSCLC cells. Moreover, SLC7A5 overexpression abolished the inhibitory effects of exosomal METTL3-decreased CAFs on NSCLC cells. In addition, METTL3 inhibition in CAF exosomes impeded NSCLC growth in vivo. In all, CAFs-derived exosomal METTL3 promoted the proliferation, invasion, stemness and glutaminolysis in NSCLC cells by inducing SLC7A5 m6A modification. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. UPLC-Q-TOF/MS-Based Serum Metabolomics Reveals Potential Anti-tumor Mechanism of Banxia Xiexin Decoction in Colorectal Cancer Mice.
- Author
-
Yue, Yin-zi, Li, Ming-xuan, Wang, Xiao-hui, Qin, Yuan-yuan, Wang, Ya-hui, Tan, Jin-hua, Su, Lian-lin, and Yan, Shuai
- Subjects
ARGININE metabolism ,THERAPEUTIC use of antineoplastic agents ,GLUTAMINE metabolism ,INFLAMMATION prevention ,GLUTAMIC acid metabolism ,TRYPTOPHAN metabolism ,PHENYLALANINE metabolism ,TYROSINE metabolism ,NITROGEN metabolism ,PREVENTION of weight loss ,CHINESE medicine ,BIOLOGICAL models ,MESALAMINE ,LIQUID chromatography-mass spectrometry ,HERBAL medicine ,ANTINEOPLASTIC agents ,ENZYME-linked immunosorbent assay ,COLORECTAL cancer ,TUMOR markers ,MICE ,METABOLITES ,GENE expression ,ANIMAL experimentation ,METABOLISM ,METABOLOMICS ,CYTOKINES ,IMMUNITY ,DRUG dosage ,THERAPEUTICS ,PHARMACODYNAMICS ,DRUG administration - Abstract
Objective: To clarify the potential mechanism of Banxia Xiexin Decoction (BXD) on colorectal cancer (CRC) from the perspective of metabolomics. Methods: Forty male C57BL/6 mice were randomly divided into normal control (NC), azoxymethane/dextran sulfate sodium (AOM/DSS) model, low-dose BXD (L-BXD), high-dose BXD (H-BXD) and mesalamine (MS) groups according to a random number table, 8 mice in each group. Colorectal cancer model was induced by AOM/DSS. BXD was administered daily at doses of 3.915 (L-BXD) and 15.66 g/kg (H-BXD) by gavage for consecutive 21 days, and 100 mg/kg MS was used as positive control. Following the entire modeling cycle, colon length of mice was measured and quantity of colorectal tumors were counted. The spleen and thymus index were determined by calculating the spleen/thymus weight to body weight. Inflammatory cytokine and changes of serum metabolites were analyzed by enzyme-linked immunosorbent assay kits and ultra performance liquid chromatography-quadrupole/time-of-flight mass spectrometry (UPLC-Q/TOF-MS), respectively. Results: Notably, BXD supplementation protected against weight loss, mitigated tumor formation, and diminished histologic damage in mice treated with AOM/DSS (P<0.05 or P<0.01). Moreover, BXD suppressed expression of serum inflammatory enzymes, and improved the spleen and thymus index (P<0.05). Compared with the normal group, 102 kinds of differential metabolites were screened in the AOM/DSS group, including 48 potential biomarkers, involving 18 main metabolic pathways. Totally 18 potential biomarkers related to CRC were identified, and the anti-CRC mechanism of BXD was closely related to D-glutamine and D-glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, arginine biosynthesis, nitrogen metabolism and so on. Conclusion: BXD exerts partial protective effects on AOM/DSS-induced CRC by reducing inflammation, protecting organism immunity ability, and regulating amino acid metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Tripartite Motif-Containing 2, a Glutamine Metabolism-Associated Protein, Predicts Poor Patient Outcome in Triple-Negative Breast Cancer Treated with Chemotherapy.
- Author
-
Masisi, Brendah K., El Ansari, Rokaya, Alfarsi, Lutfi, Fakroun, Ali, Erkan, Busra, Ibrahim, Asmaa, Toss, Michael, Ellis, Ian O., Rakha, Emad A., and Green, Andrew R.
- Subjects
- *
BREAST cancer prognosis , *GLUTAMINE metabolism , *PROTEIN metabolism , *CANCER invasiveness , *BREAST tumors , *CELL physiology , *TREATMENT effectiveness , *TRANSCRIPTION factors , *DESCRIPTIVE statistics , *CANCER chemotherapy , *GENE expression , *MESSENGER RNA , *IMMUNOHISTOCHEMISTRY , *METASTASIS , *TUMOR classification , *SEQUENCE analysis - Abstract
Simple Summary: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with limited treatment options. This study looked for markers in breast cancer cells linked to glutamine metabolism, an important fuel source for cancer cells. It found a protein called TRIM2 to be highly expressed alongside a key glutamine metabolism enzyme in TNBC. High levels of TRIM2 were linked to a worse chance of spread of cancer to distant sites. This association was particularly strong in patients who received chemotherapy. These findings suggest TRIM2 could be a valuable prognostic marker for TNBC patients, especially those undergoing chemotherapy. Further research is needed to understand how TRIM2 functions and its connection to glutamine metabolism in breast cancer. Background: Breast cancer (BC) remains heterogeneous in terms of prognosis and response to treatment. Metabolic reprogramming is a critical part of oncogenesis and a potential therapeutic target. Glutaminase (GLS), which generates glutamate from glutamine, plays a role in triple-negative breast cancer (TNBC). However, targeting GLS directly may be difficult, as it is essential for normal cell function. This study aimed to determine potential targets in BC associated with glutamine metabolism and evaluate their prognostic value in BC. Methods: The iNET model was used to identify genes in BC that are associated with GLS using RNA-sequencing data. The prognostic significance of tripartite motif-containing 2 (TRIM2) mRNA was assessed in BC transcriptomic data (n = 16,575), and TRIM2 protein expression was evaluated using immunohistochemistry (n = 749) in patients with early-stage invasive breast cancer with long-term follow-up. The associations between TRIM2 expression and clinicopathological features and patient outcomes were evaluated. Results: Pathway analysis identified TRIM2 expression as an important gene co-expressed with high GLS expression in BC. High TRIM2 mRNA and TRIM2 protein expression were associated with TNBC (p < 0.01). TRIM2 was a predictor of poor distant metastasis-free survival (DMFS) in TNBC (p < 0.01), and this was independent of established prognostic factors (p < 0.05), particularly in those who received chemotherapy (p < 0.05). In addition, TRIM2 was a predictor of shorter DMFS in TNBC treated with chemotherapy (p < 0.01). Conclusions: This study provides evidence of an association between TRIM2 and poor patient outcomes in TNBC, especially those treated with chemotherapy. The molecular mechanisms and functional behaviour of TRIM2 and the functional link with GLS in BC warrant further exploration using in vitro models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. Inhibition of asparagine synthetase effectively retards polycystic kidney disease progression.
- Author
-
Clerici, Sara, Podrini, Christine, Stefanoni, Davide, Distefano, Gianfranco, Cassina, Laura, Steidl, Maria Elena, Tronci, Laura, Canu, Tamara, Chiaravalli, Marco, Spies, Daniel, Bell 3rd, Thomas A, Costa, Ana SH, Esposito, Antonio, D'Alessandro, Angelo, Frezza, Christian, Bachi, Angela, and Boletta, Alessandra
- Abstract
Polycystic kidney disease (PKD) is a genetic disorder characterized by bilateral cyst formation. We showed that PKD cells and kidneys display metabolic alterations, including the Warburg effect and glutaminolysis, sustained in vitro by the enzyme asparagine synthetase (ASNS). Here, we used antisense oligonucleotides (ASO) against Asns in orthologous and slowly progressive PKD murine models and show that treatment leads to a drastic reduction of total kidney volume (measured by MRI) and a prominent rescue of renal function in the mouse. Mechanistically, the upregulation of an ATF4–ASNS axis in PKD is driven by the amino acid response (AAR) branch of the integrated stress response (ISR). Metabolic profiling of PKD or control kidneys treated with Asns-ASO or Scr-ASO revealed major changes in the mutants, several of which are rescued by Asns silencing in vivo. Indeed, ASNS drives glutamine-dependent de novo pyrimidine synthesis and proliferation in cystic epithelia. Notably, while several metabolic pathways were completely corrected by Asns-ASO, glycolysis was only partially restored. Accordingly, combining the glycolytic inhibitor 2DG with Asns-ASO further improved efficacy. Our studies identify a new therapeutic target and novel metabolic vulnerabilities in PKD. Synopsis: Metabolic Reprogramming, such as glycolysis and glutaminolysis, are key features of polycystic kidney disease (PKD). Asparagine synthetase drives glutamine utilization in this disease and it is upregulated in human and mouse tissues. Its inhibition retards disease progression and rescues metabolic derangement in PKD mice. Glutamine utilization and cyst expansion are driven by ASNS upregulation in PKD. PKD cystic phenotype and metabolic rewiring are hampered by Asns-ASO. Glutaminolysis, pyrimidine biosynthesis and proliferation are driven by a GCN2-ATF4-ASNS axis. PKD is further delayed by co-targeting glutaminolysis and glycolysis with Asns-ASO and 2DG. Metabolic Reprogramming, such as glycolysis and glutaminolysis, are key features of polycystic kidney disease (PKD). Asparagine synthetase drives glutamine utilization in this disease and it is upregulated in human and mouse tissues. Its inhibition retards disease progression and rescues metabolic derangement in PKD mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Glutamine Metabolism Heterogeneity in Glioblastoma Unveils an Innovative Combination Therapy Strategy.
- Author
-
Fu, Huangde, Wu, Shengtian, Shen, Hechun, Luo, Kai, Huang, Zhongxiang, Lu, Nankun, Li, Yaolin, Lan, Qian, and Xian, Yishun
- Abstract
Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism–based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.