11,009 results on '"Glutamine metabolism"'
Search Results
152. The Heterogeneity of Liver Cancer Metabolism
- Author
-
Salazar, Javier, Le, Anne, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Xiao, Junjie, Series Editor, and Le, Anne, editor
- Published
- 2021
- Full Text
- View/download PDF
153. The Intricate Metabolism of Pancreatic Cancers
- Author
-
Camelo, Felipe, Le, Anne, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Xiao, Junjie, Series Editor, and Le, Anne, editor
- Published
- 2021
- Full Text
- View/download PDF
154. The Multifaceted Glioblastoma: From Genomic Alterations to Metabolic Adaptations
- Author
-
Quinones, Addison, Le, Anne, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Xiao, Junjie, Series Editor, and Le, Anne, editor
- Published
- 2021
- Full Text
- View/download PDF
155. Glutamine Metabolism in Cancer
- Author
-
Li, Ting, Copeland, Christopher, Le, Anne, Crusio, Wim E., Series Editor, Dong, Haidong, Series Editor, Radeke, Heinfried H., Series Editor, Rezaei, Nima, Series Editor, Xiao, Junjie, Series Editor, and Le, Anne, editor
- Published
- 2021
- Full Text
- View/download PDF
156. Baicalein induces apoptosis by inhibiting the glutamine-mTOR metabolic pathway in lung cancer.
- Author
-
Li J, Zhang D, Wang S, Yu P, Sun J, Zhang Y, Meng X, Li J, and Xiang L
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, A549 Cells, Signal Transduction drug effects, Mice, Nude, Glutaminase metabolism, Metabolic Networks and Pathways drug effects, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung metabolism, Molecular Docking Simulation, Male, Scutellaria baicalensis, Flavanones pharmacology, TOR Serine-Threonine Kinases metabolism, Glutamine metabolism, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Apoptosis drug effects, Cell Proliferation drug effects, Xenograft Model Antitumor Assays
- Abstract
Introduction: Baicalein, a bioactive component of Scutellaria baicalensis Georgi, has been shown to promote apoptosis in non-small cell lung cancer cells. However, previous studies have not determined if baicalein exerts proapoptotic effects by modulating the metabolic pathways., Objective: To investigate if baicalein induces apoptosis in lung cancer cells by modulating the glutamine-mTOR metabolic pathway., Methods: The in vivo anti-lung cancer activity of baicalein (50, 100, and 200 mg/kg) was evaluated using a xenograft model. In vitro experiments were used to assess the efficacy of baicalein (for H1299: 12.5, 25, and 50 μM; for A549: 10, 20, and 40 μM) on lung cancer cell proliferation, colony formation, and apoptosis. Metabolomics analysis was performed using liquid chromatography-mass spectrometry. The binding of baicalein to glutamine transporters and glutaminase was examined using molecular docking. The overexpression of glutamine transporters was validated using qRT-PCR and western blot analyses. The levels of ASCT2, LAT1, GLS1, p-mTOR, mTOR, and apoptosis-related proteins were evaluated using western blot analysis., Results: Baicalein inhibited lung cancer xenograft tumor growth in vivo and suppressed proliferation and promoted apoptosis in lung cancer cells in vitro. Additionally, baicalein altered amino acid metabolites, especially glutamine metabolites, in H1299 and A549 cells. Mechanistically, baicalein interacted with glutamine transporters as well as glutaminase and inhibited their activation. The expression of mTOR, an apoptosis-related protein and downstream target of glutamine metabolism, was also inhibited by baicalein treatment. Importantly, we next demonstrated the suppression of mTOR signaling and the induction of apoptosis by baicalein were achieved by regulating glutamine metabolism., Conclusion: Baicalein inhibited the mTOR signaling pathway and induced apoptosis by downregulating glutamine metabolism. The potential of baicalein to induce apoptosis in lung cancer cells by selectively targeting the glutamine-mTOR pathway suggests an encouraging approach for treating lung cancer., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023. Published by Elsevier B.V.)
- Published
- 2025
- Full Text
- View/download PDF
157. Targeting glutamine metabolism crosstalk with tumor immune response.
- Author
-
Dong C, Zhao Y, Han Y, Li M, and Wang G
- Subjects
- Humans, Animals, Glutamine metabolism, Neoplasms immunology, Neoplasms metabolism, Neoplasms drug therapy, Neoplasms pathology, Tumor Microenvironment immunology, Immunotherapy methods
- Abstract
Glutamine, akin to glucose, is a fundamental nutrient for human physiology. Tumor progression is often accompanied by elevated glutamine consumption, resulting in a disrupted nutritional balance and metabolic reprogramming within the tumor microenvironment. Furthermore, immune cells, which depend on glutamine for metabolic support, may experience functional impairments and dysregulation. Although the role of glutamine in tumors has been extensively studied, the specific impact of glutamine competition on immune responses, as well as the precise cellular alterations within immune cells, remains incompletely understood. In this review, we summarize the consequences of glutamine deprivation induced by tumor-driven glutamine uptake on immune cells, assessing the underlying mechanisms from the perspective of various components of the immune microenvironment. Additionally, we discuss the potential synergistic effects of glutamine supplementation and immunotherapy, offering insights into future research directions. This review provides compelling evidence for the integration of glutamine metabolism and immunotherapy as a promising strategy in cancer therapy., Competing Interests: Declaration of competing interest The authors declare that they have no competing interests., (Copyright © 2025 Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
158. Plant organic nitrogen nutrition: costs, benefits, and carbon use efficiency.
- Author
-
Tünnermann L, Aguetoni Cambui C, Franklin O, Merkel P, Näsholm T, and Gratz R
- Subjects
- Glutamine metabolism, Plant Shoots growth & development, Plant Shoots metabolism, Nitrogen Isotopes, Nitrates metabolism, Carbon metabolism, Nitrogen metabolism, Plant Roots metabolism, Plant Roots growth & development, Arabidopsis growth & development, Arabidopsis metabolism, Biomass
- Abstract
Differences in soil mobility and assimilation costs between organic and inorganic nitrogen (N) compounds would hypothetically induce plant phenotypic plasticity to optimize acquisition of, and performance on, the different N forms. Here we evaluated this hypothesis experimentally and theoretically. We grew Arabidopsis in split-root setups combined with stable isotope labelling to study uptake and distribution of carbon (C) and N from l-glutamine (l-gln) and NO
3 - and assessed the effect of the N source on biomass partitioning and carbon use efficiency (CUE). Analyses of stable isotopes showed that 40-48% of C acquired from l-gln resided in plants, contributing 7-8% to total C of both shoots and roots. Plants grown on l-gln exhibited increased root mass fraction and root hair length and a significantly lower N uptake rate per unit root biomass but displayed significantly enhanced CUE. Our data suggests that organic N nutrition is linked to a particular phenotype with extensive growth of roots and root hairs that optimizes for uptake of less mobile N forms. Increased CUE and lower N uptake per unit root growth may be key facets linked to the organic N phenotype., (© 2024 The Author(s). New Phytologist © 2024 New Phytologist Foundation.)- Published
- 2025
- Full Text
- View/download PDF
159. Addressing Clinical Limitations of Glutaminase Inhibitors: Novel Strategies for Osimertinib-Resistant Lung Cancer by Exploiting Glutamine Metabolic Dependency.
- Author
-
Huang J, Zhang X, Zhang H, Li Y, Huang H, Li Z, Qiu Z, Wu H, Huang D, Xu X, and Bian J
- Subjects
- Humans, Mice, Animals, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung genetics, Disease Models, Animal, Prodrugs pharmacology, Indoles, Pyrimidines, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lung Neoplasms genetics, Glutaminase antagonists & inhibitors, Glutaminase metabolism, Glutaminase genetics, Glutamine metabolism, Acrylamides pharmacology, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Aniline Compounds pharmacology, Aniline Compounds therapeutic use
- Abstract
Overcoming acquired resistance to Osimertinib remains a critical challenge in treating NSCLC. This research indicates that Osimertinib-resistant cells exhibit a strong dependence on glutamine metabolism. However, targeting GLS1 shows limited anticancer effects, probably because it cannot fully block the glutamine metabolic pathway. The investigation reveals that a more effective strategy involves simultaneously inhibiting both ASCT2 and GLS1. After confirming the efficacy of this dual-targeting approach against Osimertinib-resistant cells in preclinical models, the potential of utilizing a broad-spectrum glutamine metabolism antagonist is further explored to achieve superior antitumor efficacy. DON, broad-spectrum glutamine antagonist, presents toxicity issues. Herein, the high NQO1 expression in Osimertinib-resistant NSCLC cells is leveraged to design an NQO1-responsive DON prodrug, 10e (LBJ-10e). This prodrug demonstrates superior safety compared to natural DON and greater antitumor activity against resistant tumors compared to the clinical phase II drug DRP104. These findings may address the clinical limitations of GLS1 allosteric inhibitors and underscore prodrug strategies in effectively treating Osimertinib-resistant lung cancer, providing a foundation for future clinical trials., (© 2024 The Author(s). Advanced Science published by Wiley‐VCH GmbH.)
- Published
- 2025
- Full Text
- View/download PDF
160. Ambient PM 2.5 orchestrates M1 polarization of alveolar macrophages via activating glutaminase 1-mediated glutaminolysis in acute lung injury.
- Author
-
Su J, Tu Y, Hu X, Wang Y, Chen M, Cao X, Shao M, Zhang F, and Ding W
- Subjects
- Animals, Mice, Glutamine metabolism, Mice, Inbred C57BL, Male, NF-kappa B metabolism, China, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Acute Lung Injury chemically induced, Acute Lung Injury metabolism, Glutaminase metabolism, Macrophages, Alveolar drug effects, Macrophages, Alveolar metabolism, Particulate Matter toxicity, Air Pollutants toxicity
- Abstract
The temporary explosive growth events of atmospheric fine particulate matter (PM
2.5 ) pollution during late autumn and winter seasons still frequently occur in China. High-concentration exposure to PM2.5 aggravates lung inflammation, leading to acute lung injury (ALI). Alveolar macrophages (AMs) participate in PM2.5 -induced pulmonary inflammation and injury. The polarization of AMs is dependent on metabolic reprogramming. However, the mechanism underlying the PM2.5 -induced glutaminase-mediated glutaminolysis in AM polarization is still largely obscure. In this study, we found that PM2.5 -treated mice exhibited pulmonary dysfunction and inflammation. The concentrations of glutamate and succinate were increased in PM2.5 -treated lungs and AMs compared with the controls, whereas glutamine and α-ketoglutarate (α-KG) levels were decreased, indicating that glutaminolysis in AMs was aberrantly activated as evidenced by increased mRNA and protein levels of GLS1 after PM2.5 exposure. Moreover, we determined that the GLS1/nuclear factor kappa-B (NF-κB)/hypoxia-inducible factor-1α (HIF-1α) pathway regulated M1 polarization of AMs upon PM2.5 exposure. Inhibition of glutaminolysis by GLS1 specific inhibitor CB-839 and GLS1 siRNA significantly decreased PM2.5 -induced M1 macrophage polarization and attenuated pulmonary damage. Taken together, our findings reveal a novel mechanism by which a metabolic program regulates M1 polarization of AMs and suggest that GLS1-mediated glutaminolysis is a potential therapeutic target for treating PM2.5 -induced ALI., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
161. Examination of methods to separate overlapping metabolites at 7T.
- Author
-
Bell TK, Goerzen D, Near J, and Harris AD
- Subjects
- Humans, Male, Female, Reproducibility of Results, Adult, Glutamic Acid metabolism, Creatine metabolism, Choline metabolism, Young Adult, Glutamine metabolism, Aspartic Acid analogs & derivatives, Aspartic Acid metabolism, Algorithms, Brain diagnostic imaging, Brain metabolism, Magnetic Resonance Spectroscopy methods
- Abstract
Purpose: Neurochemicals of interest quantified by MRS are often composites of overlapping signals. At higher field strengths (i.e., 7T), there is better separation of these signals. As the availability of higher field strengths is increasing, it is important to re-evaluate the separability of overlapping metabolite signals., Methods: This study compares the ability of stimulated echo acquisition mode (STEAM-8; TE = 8 ms), short-TE semi-LASER (sLASER-34; TE = 34 ms), and long-TE semi-LASER (sLASER-105; TE = 105 ms) acquisitions to separate the commonly acquired neurochemicals at 7T (Glx, consisting of glutamate and glutamine; total N-acetyl aspartate, consisting of N-acetyl aspartate and N-acetylaspartylglutamate; total creatine, consisting of creatine and phosphocreatine; and total choline, consisting of choline, phosphocholine, and glycerophosphocholine)., Results: sLASER-34 produced the lowest fit errors for most neurochemicals; however, STEAM-8 had better within-subject reproducibility and required fewer subjects to detect a change between groups. However, this is dependent on the neurochemical of interest., Conclusion: We recommend short-TE STEAM for separation of most standard neurochemicals at 7T over short-TE or long-TE sLASER., (© 2024 The Author(s). Magnetic Resonance in Medicine published by Wiley Periodicals LLC on behalf of International Society for Magnetic Resonance in Medicine.)
- Published
- 2025
- Full Text
- View/download PDF
162. Elevated posterior insula glutamate in patients with sickle cell disease.
- Author
-
Zhou X, Ichesco E, Pucka AQ, Liu Z, O'Brien AR, Harte SE, Harris RE, and Wang Y
- Subjects
- Humans, Female, Male, Adult, Young Adult, Proton Magnetic Resonance Spectroscopy, Glutamine metabolism, Middle Aged, Cerebral Cortex metabolism, Cerebral Cortex diagnostic imaging, Anemia, Sickle Cell metabolism, Anemia, Sickle Cell complications, Glutamic Acid metabolism, Insular Cortex metabolism, Insular Cortex diagnostic imaging
- Abstract
Sickle cell disease (SCD) is an inherited hemolytic disorder accompanied by chronic pain and recurrent acute painful episodes known as vaso-occlusive crises (VOCs). Increased Glx (glutamate+glutamine) and lowered GABA concentration have been reported in the insula of patients with fibromyalgia, a nociplastic chronic pain condition, and may affect the pathophysiology of pain-related syndromes.Therefore, proton magnetic resonance spectroscopy (
1 H-MRS) was conducted to measure levels of Glx and other brain metabolites using a single voxel (size: 2×3×3 cm3 ) in the right posterior insula cortex (PIC) in 17 individuals with SCD and 17 ethnicity-, age- and sex-matched healthy controls (HCs). The frequency of VOCs in the preceding 12 months was recorded. The concentration of Glx (p=0.019) and the ratio of Glx to tCr (total creatine, p=0.035) in the PIC were significantly higher in patients with SCD as compared to matched HCs (n=17). Secondary analyses with the unpaired full sample of 24 SCD also showed a significantly higher level of Glx/tCr than HCs (n=19), with a positive correlation between the level of Glx/tCr and the number of VOCs (p=0.034, r=0.476), as well as a negative correlation between Glx and sensory sensitivity assessed by tonic pressure pain in gastrocnemius area of the non-dominant leg (p=0.040, r=-0.462). The unpaired full sample additionally revealed a significant difference in sensory sensitivity (p=0.050). Altered metabolites such as GABA and myo-inositol were also observed between SCD and HCs. These results suggest that elevated excitatory neurotransmission in the insula might contribute to nociplastic pain in SCD. PERSPECTIVE: Our work highlighted the innovative finding of elevated levels of the excitatory neurotransmitter glutamate with glutamine in patients with SCD compared to healthy controls. The positive relationship between Glx/tCr and the frequency of VOCs suggests that an excitatory brain neurotransmitter imbalance may be involved in VOCs., Competing Interests: Declaration of Competing Interest The remaining authors declare no competing interests., (Copyright © 2025 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
163. Alzheimer's disease: an integrative bioinformatics and machine learning analysis reveals glutamine metabolism-associated gene biomarkers.
- Author
-
Xing N, Yan J, Gao R, Zhang A, He H, Zheng M, and Li G
- Subjects
- Humans, Alzheimer Disease genetics, Alzheimer Disease metabolism, Glutamine metabolism, Glutamine genetics, Machine Learning, Computational Biology methods, Biomarkers metabolism
- Abstract
Background: Alzheimer's disease (AD), a hallmark of age-related cognitive decline, is defined by its unique neuropathology. Metabolic dysregulation, particularly involving glutamine (Gln) metabolism, has emerged as a critical but underexplored aspect of AD pathophysiology, representing a significant gap in our current understanding of the disease., Methods: To investigate the involvement of GlnMgs in AD, we conducted a comprehensive bioinformatic analysis. We began by identifying differentially expressed GlnMgs from a curated list of 34 candidate genes. Subsequently, we employed GSEA and GSVA to assess the biological significance of these GlnMgs. Advanced techniques such as Lasso regression and SVM-RFE were utilized to identify key hub genes and evaluate the diagnostic potential of 14 central GlnMgs in AD. Additionally, we examined their correlations with clinical parameters and validated their expression across multiple independent AD cohorts (GSE5281, GSE37263, GSE106241, GSE132903, GSE63060)., Results: Our rigorous analysis identified 14 GlnMgs-GLS2, GLS, GLUD2, GLUL, GOT1, HAL, AADAT, PFAS, ASNSD1, PPAT, NIT2, ALDH5A1, ASRGL1, and ATCAY-as potential contributors to AD pathogenesis. These genes were implicated in vital biological processes, including lipid transport and the metabolism of purine-containing compounds, in response to nutrient availability. Notably, these GlnMgs demonstrated significant diagnostic potential, highlighting their utility as both diagnostic and prognostic biomarkers for AD., Conclusions: Our study uncovers 14 GlnMgs with potential links to AD, expanding our understanding of the disease's molecular underpinnings and offering promising avenues for biomarker development. These findings not only enhance the molecular landscape of AD but also pave the way for future diagnostic and therapeutic innovations, potentially reshaping AD diagnostics and patient care., Competing Interests: Declarations. Ethics approval and consent to participation: This manuscript is not a clinical trial, hence the ethics approval and consent to participation are not applicable. Consent for publication: All authors have read and approved this manuscript to be considered for publication. Competing interests: The authors declare no competing interests., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
164. Evolution Enhances Kemp Eliminase Activity by Optimizing Oxyanion Stabilization and Conformational Flexibility.
- Author
-
Doustmohammadi H, Sanchez J, Ram Mahato D, and Osuna S
- Subjects
- Water chemistry, Anions chemistry, Glutamine chemistry, Glutamine metabolism, Mercury chemistry, Protein Conformation, Biocatalysis, Mutation, Molecular Dynamics Simulation, Catalytic Domain
- Abstract
The base-promoted Kemp elimination reaction has been used as a model system for enzyme design. Among the multiple computationally designed and evolved Kemp eliminases generated along the years, the HG3-to-HG3.17 evolutionary trajectory is particularly interesting due to the high catalytic efficiency of HG3.17 and the debated role of glutamine 50 (Gln50) as potential oxyanion stabilizer. This study aims to elucidate the structural and dynamic changes along the evolutionary pathway from HG3 to HG3.17 that contribute to improved catalytic efficiency. In particular, we evaluate key variants along the HG3 evolutionary trajectory via molecular dynamics simulations coupled to non-covalent interactions and water analysis. Our computational study indicates that HG3.17 can adopt a catalytically competent conformation promoted by a water-mediated network of non-covalent interactions, in which aspartate 127 (Asp127) is properly positioned for proton abstraction and Gln50 and to some extent mutation cysteine 84 (Cys84) contribute to oxyanion stabilization. We find that HG3.17 exhibits a rather high flexibility of Gln50, which is regulated by the conformation adopted by the active site residue tryptophan 44 (Trp44). This interplay between Gln50 and Trp44 positioning induced by distal active site mutations affects the water-mediated network of non-covalent interactions, Gln50 preorganization, and water content of the active site pocket., (© 2024 The Author(s). Chemistry - A European Journal published by Wiley-VCH GmbH.)
- Published
- 2025
- Full Text
- View/download PDF
165. Significance of birth in the maintenance of quiescent neural stem cells.
- Author
-
Kawase K, Nakamura Y, Wolbeck L, Takemura S, Zaitsu K, Ando T, Jinnou H, Sawada M, Nakajima C, Rydbirk R, Gokenya S, Ito A, Fujiyama H, Saito A, Iguchi A, Kratimenos P, Ishibashi N, Gallo V, Iwata O, Saitoh S, Khodosevich K, and Sawamoto K
- Subjects
- Animals, Mice, Glutamine metabolism, Female, Animals, Newborn, Ependymoglial Cells metabolism, Ependymoglial Cells cytology, Parturition, Cell Proliferation, Brain metabolism, Brain cytology, Neural Stem Cells metabolism, Neural Stem Cells cytology, Neurogenesis
- Abstract
Birth is one of the most important life events for animals. However, its significance in the developmental process is not fully understood. Here, we found that birth-induced alteration of glutamine metabolism in radial glia (RG), the embryonic neural stem cells (NSCs), is required for the acquisition of quiescence and long-term maintenance of postnatal NSCs. Preterm birth impairs this cellular process, leading to transient hyperactivation of RG. Consequently, in the postnatal brain, the NSC pool is depleted and neurogenesis is decreased. We also found that the maintenance of quiescent RG after preterm birth improves postnatal neurogenesis. This study demonstrates the significance of birth in the maintenance of quiescent NSCs.
- Published
- 2025
- Full Text
- View/download PDF
166. Identification of glutamine as a potential therapeutic target in dry eye disease.
- Author
-
Chen X, Zhang C, Peng F, Wu L, Zhuo D, Wang L, Zhang M, Li Z, Tian L, Jie Y, Huang Y, Yang X, Li X, Lei F, and Cheng Y
- Subjects
- Humans, Animals, Glutaminase genetics, Glutaminase antagonists & inhibitors, Glutaminase metabolism, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells drug effects, Mice, Male, Female, Rats, Disease Models, Animal, Mesenchymal Stem Cell Transplantation, Thymosin, Glutamine metabolism, Dry Eye Syndromes drug therapy, Dry Eye Syndromes genetics, Dry Eye Syndromes pathology
- Abstract
Dry eye disease (DED) is a prevalent inflammatory condition significantly impacting quality of life, yet lacks effective pharmacological therapies. Herein, we proposed a novel approach to modulate the inflammation through metabolic remodeling, thus promoting dry eye recovery. Our study demonstrated that co-treatment with mesenchymal stem cells (MSCs) and thymosin beta-4 (Tβ4) yielded the best therapeutic outcome against dry eye, surpassing monotherapy outcomes. In situ metabolomics through matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) revealed increased glutamine levels in cornea following MSC + Tβ4 combined therapy. Inhibition of glutamine reversed the anti-inflammatory, anti-apoptotic, and homeostasis-preserving effects observed with combined therapy, highlighting the critical role of glutamine in dry eye therapy. Clinical cases and rodent model showed elevated expression of glutaminase (GLS1), an upstream enzyme in glutamine metabolism, following dry eye injury. Mechanistic studies indicated that overexpression and inhibition of GLS1 counteracted and enhanced, respectively, the anti-inflammatory effects of combined therapy, underscoring GLS1's pivotal role in regulating glutamine metabolism. Furthermore, single-cell sequencing revealed a distinct subset of pro-inflammatory and pro-fibrotic corneal epithelial cells in the dry eye model, while glutamine treatment downregulated those subclusters, thereby reducing their inflammatory cytokine secretion. In summary, glutamine effectively ameliorated inflammation and the occurrence of apoptosis by downregulating the pro-inflammatory and pro-fibrotic corneal epithelial cells subclusters and the related IκBα/NF-κB signaling. The present study suggests that glutamine metabolism plays a critical, previously unrecognized role in DED and proposes an attractive strategy to enhance glutamine metabolism by inhibiting the enzyme GLS1 and thus alleviating inflammation-driven DED progression., Competing Interests: Competing interests: The authors declare no competing interests., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
167. Targeting Glutamine Metabolic Reprogramming in Pancreatic Cancer: Current Insights and Future Directions.
- Author
-
Ma Y, Wang M, Zhang K, Ren F, Wang Y, Zhang W, Kan C, Han F, Sun H, and Sun X
- Abstract
Pancreatic Cancer (PC) is a devastating malignancy with a poor prognosis and in-creasing morbidity. Current treatment strategies have limited efficacy in improving patient survival. Metabolic reprogramming is a hallmark of cancer and plays a key role in the pro-gression and maintenance of PC. PC cells exhibit a unique glutamine metabolism that is dis-tinct from other cancer types. The non-classical pathway of glutamine metabolic reprogram-ming plays a "permissive role" in the survival and proliferation of PC cells, mainly by affect-ing the redox homeostasis of the cells. In this review, we compare and contrast the canonical and non-canonical glutamine metabolic pathways and highlight recent advances in targeting non-canonical glutamine metabolism for therapeutic intervention. This may provide novel in-sights and opportunities for exploiting glutamine metabolic reprogramming in PC treatment., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2025
- Full Text
- View/download PDF
168. SLC1A5 is a key regulator of glutamine metabolism and a prognostic marker for aggressive luminal breast cancer.
- Author
-
Alfarsi LH, Ansari RE, Erkan B, Fakroun A, Craze ML, Aleskandarany MA, Cheng KW, Ellis IO, Rakha EA, and Green AR
- Subjects
- Humans, Female, Prognosis, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Middle Aged, Cell Proliferation, Aged, Amino Acid Transport System ASC metabolism, Amino Acid Transport System ASC genetics, Glutamine metabolism, Minor Histocompatibility Antigens metabolism, Minor Histocompatibility Antigens genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms genetics, Breast Neoplasms mortality, Biomarkers, Tumor metabolism, Biomarkers, Tumor genetics
- Abstract
Cancer cells exhibit altered metabolism, often relying on glutamine (Gln) for growth. Breast cancer (BC) is a heterogeneous disease with varying clinical outcomes. We investigated the role of the amino acid transporter SLC1A5 (ASCT2) and its association with BC subtypes and patient outcomes. In large BC cohorts, SLC1A5 mRNA (n = 9488) and SLC1A5 protein (n = 1274) levels were assessed and correlated their expression with clinicopathological features, molecular subtypes, and patient outcomes. In vitro SLC1A5 knockdown and inhibition studies in luminal BC cell lines (ZR-75-1 and HCC1500) were used to further explore the role of SLC1A5 in Gln metabolism. Statistical analysis was performed using chi-squared tests, ANOVA, Spearman's correlation, Kaplan-Meier analysis, and Cox regression. SLC1A5 mRNA and SLC1A5 protein expression were strongly correlated in luminal B, HER2 + and triple-negative BC (TNBC). Both high SLC1A5 mRNA and SLC1A5 protein expression were associated with larger tumour size, higher grade, and positive axillary lymph node metastases (P < 0.01). Importantly, high SLC1A5 expression correlated with poor BC-specific survival specifically in the highly proliferative luminal subtype (P < 0.001). Furthermore, SLC1A5 knockdown by siRNA or GPNA inhibition significantly reduced cell proliferation and glutamine uptake in ZR-75-1 cells. Our findings suggest SLC1A5 plays a key role in the aggressive luminal BC subtype and represents a potential therapeutic target. Further research is needed to explore SLC1A5 function in luminal BC and its association with Gln metabolism pathways., Competing Interests: Declarations. Competing interests: The authors declare no competing interests. Ethical approval: This article does not contain any studies with human participants or animals performed This work was performed according to REMARK guidelines or tumour prognostic study and obtained ethics approval by the North West–Greater Manchester Central Research Ethics Committee under the title: Nottingham Health Science Biobank (NHSB), reference number 15/NW/0685. We can declare that this study is complying with Helsinki declaration., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
169. GlutaR: A High-Performance Fluorescent Protein-Based Sensor for Spatiotemporal Monitoring of Glutamine Dynamics In Vivo.
- Author
-
Liu B, Zhao Z, Wang P, Aihemaiti K, Zhu L, Wei Q, Li W, Yuan X, Wu J, Jiang C, Hao M, and Wang J
- Subjects
- Humans, Animals, HeLa Cells, Mice, Luminescent Proteins metabolism, Luminescent Proteins chemistry, Fluorescent Dyes chemistry, Glutamate-Ammonia Ligase metabolism, Glutamine metabolism, Glutamine chemistry
- Abstract
Glutamine is the most abundant amino acid in human blood and muscle, and is integral to a wide variety of functions in cancer cells. However, the inability to monitor the subcellular distribution of glutamine in real-time has obscured understanding of glutamine metabolism under physiological and pathological conditions. Here, we report the development of a genetically encoded fluorescent sensor and demonstrate how this GlnBP-cpYFP fusion "GlutaR sensor" undergoes glutamine-induced conformational changes reflected in detectable fluorescence responses. Obtained after iterative screening of approximately 1,600 variants, GlutaR exhibits a ratiometric readout, fast response kinetics, and high responsivity (R
488/405 of ~1000 %), and we demonstrate its selectivity for monitoring glutamine fluctuations in multiple cell types. Additionally, using digitonin permeabilization of GlutaR HeLa cells, we generated a calibration curve and performed in situ titration to quantify free glutamine concentrations in subcellular compartments (cytosol, nucleus, mitochondria). Subsequently, we applied GlutaR to investigate how chemical and genetic inhibition of glutamine synthetase (GS) and glutaminase (GLS) differentially alter glutamine levels in subcellular compartments. Finally, we demonstrate GlutaR's ability to monitor dynamic glutamine levels in muscle and liver tissues of diabetic mice in vivo. These findings collectively demonstrate GlutaR as a versatile tool for the spatiotemporal characterization of glutamine metabolism in living cells and tissues., (© 2024 Wiley-VCH GmbH.)- Published
- 2025
- Full Text
- View/download PDF
170. Wall shear stress modulates metabolic pathways in endothelial cells.
- Author
-
Simões-Faria R, Daems M, Peacock HM, Declercq M, Willems A, Jones EAV, and Ghesquière B
- Subjects
- Humans, Glutamine metabolism, Metabolomics, Glutamate Dehydrogenase metabolism, Human Umbilical Vein Endothelial Cells metabolism, Cells, Cultured, Glycolysis physiology, Endothelial Cells metabolism, Stress, Mechanical, Metabolic Networks and Pathways
- Abstract
Introduction: Hemodynamic forces play a crucial role in modulating endothelial cell (EC) behavior, significantly influencing blood vessel responses. While traditional in vitro studies often explore ECs under static conditions, ECs are exposed to various hemodynamic forces in vivo. This study investigates how wall shear stress (WSS) influences EC metabolism, focusing on the interplay between WSS and key metabolic pathways., Objectives: The aim of this study is to examine the effects of WSS on EC metabolism, specifically evaluating its impact on central carbon metabolism and glycolysis using transcriptomics and tracer metabolomics approaches., Methods: ECs were exposed to WSS, and transcriptomic analysis was performed to assess gene expression changes related to metabolic pathways. Tracer metabolomics was used to track metabolic fluxes, focusing on glutamine and glycolytic metabolism. Additionally, chemical inhibition of glutamate dehydrogenase was conducted to evaluate its role in EC fitness under WSS., Results: Transcriptomic data revealed upregulation of glutamine and glutamate pathways, alongside downregulation of glycolytic activity in ECs exposed to WSS. Tracer metabolomics confirmed that WSS promotes glutamine anaplerosis into the Krebs cycle, while decreasing glycolytic metabolism. Suppression of glutamate dehydrogenase impaired EC fitness under WSS conditions., Conclusion: Our findings illuminate that ECs subjected to WSS exhibit a preference for glutamine as a key nutrient source for central carbon metabolism pathways, indicating diminished reliance on glycolysis. This study elucidates the nutritional predilections and regulatory mechanisms governing EC metabolism under WSS in vitro, underscoring the pivotal role of physical stimuli in shaping EC metabolic responses., Competing Interests: Declarations. Conflict of interest: The authors declare that they have no conflict of interest., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
171. KNTC1 introduces segmental heterogeneity to mitochondria.
- Author
-
Tsukamura A, Ariyama H, Hayashi N, Miyatake S, Okado S, Sultana S, Terakado I, Yamamoto T, Yamanaka S, Fujii S, Hamanoue H, Asano R, Mizushima T, Matsumoto N, Maruo Y, and Mori M
- Abstract
Mitochondria contribute to cellular metabolism by providing a specialised milieu for energising cells by incorporating and processing the metabolites. However, heterogeneity in the mitochondria within is only partially elucidated. Mitochondria dynamically alter their morphology and functions during the life of animals, in which cells proliferate and grow. We here show that Kntc1, a highly evolutionarily conserved protein, translocates from the Golgi apparatus to linear mitochondrial segments (LMS) upon glutamine deprivation and plays an essential role in maintaining LMS. The LMS with Kntc1 localisation exhibits an increase in the membrane potential, suggesting the role of Kntc1 in functioning as a reservoir for the energy-generating potential. Suppression of Kntc1 leads to glutamine consumption and lactate production, thus impacting cellular metabolism, eventually leading to anchorage-independent growth of cells. Indeed, the KNTC1 variant was identified in a patient with ovarian cancer, suggesting that segmental regulation of the mitochondrial function is essential for maintaining tissue integrity., (© 2025. Published by The Company of Biologists.)
- Published
- 2025
- Full Text
- View/download PDF
172. Mechanism Study of E2F8 Activation of SPC25-Mediated Glutamine Metabolism Promoting Immune Escape in Lung Adenocarcinoma.
- Author
-
Luo M, Xie L, Lin B, Su X, Liang R, Ma Z, and Li Y
- Abstract
Tumour cell immune infiltration is linked to spindle pole component 25 (SPC25). The purpose of this work was to examine the function and molecular mechanism of SPC25 in immune escape in lung adenocarcinoma (LUAD). SPC25 expression in LUAD was examined using The Cancer Genome Atlas (TCGA) database, and RT-qPCR was used to confirm the results. The study involved the use of CD8
+ T lymphocytes for immunoinfiltration analysis of SPC25, Gene Set Enrichment Analysis (GSEA) analysis of signalling pathways enriched by SPC25, identification of putative regulatory molecules of SPC25, and confirmation through the use of dual-luciferase and ChIP tests. To evaluate LUAD cell capacity for immune escape, a co-culture technique was employed. Measurements of glutamine uptake, glutamate and α-ketoglutarate levels, NADPH/NADP and GSH/GSSG ratios, and SLC1A5 expression were used to assess the levels of glutamine metabolism. LUAD had increased SPC25 expression. In LUAD cells, immune escape was facilitated by SPC25 knockdown, whereas overexpression had the reverse effect. SPC25 enrichment in the glutamine metabolism pathway was shown by GSEA analysis. Through increased glutamine metabolism brought on by SPC25 overexpression, immune escape was improved in LUAD and could be mitigated by GPNA therapy. E2F8 was also shown to be the transcription factor associated with SPC25, and they showed a binding interaction. By inhibiting glutamine metabolism through SPC25, knocking down E2F8 prevented immune escape in LUAD cells. On the other hand, the suppression of immune escape in LUAD cells caused by E2F8 knockdown was overcome by overexpression of SPC25. In LUAD, E2F8 stimulates SPC25 expression to facilitate glutamine metabolism and encourage immune escape. Our research validates a novel immune escape pathway driven by SPC25 in LUAD cells, providing LUAD patients with potentially effective immunotherapeutic approaches., (© 2025 John Wiley & Sons Ltd.)- Published
- 2025
- Full Text
- View/download PDF
173. Glutamine metabolic competition drives immunosuppressive reprogramming of intratumour GPR109A + myeloid cells to promote liver cancer progression.
- Author
-
Yang Y, Pei T, Liu C, Cao M, Hu X, Yuan J, Chen F, Guo B, Hong Y, Liu J, Li B, Li X, and Wang H
- Subjects
- Animals, Humans, Mice, Amino Acid Transport System ASC metabolism, Amino Acid Transport System ASC genetics, Myeloid Cells metabolism, Myeloid Cells immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, Myeloid-Derived Suppressor Cells metabolism, Myeloid-Derived Suppressor Cells immunology, Cell Line, Tumor, Minor Histocompatibility Antigens, Receptors, G-Protein-Coupled metabolism, Receptors, G-Protein-Coupled genetics, Glutamine metabolism, Liver Neoplasms metabolism, Liver Neoplasms pathology, Liver Neoplasms immunology, Tumor Microenvironment immunology, Disease Progression, Mice, Knockout
- Abstract
Objective: The metabolic characteristics of liver cancer drive considerable hurdles to immune cells function and cancer immunotherapy. However, how metabolic reprograming in the tumour microenvironment impairs the antitumour immune response remains unclear., Design: Human samples and multiple murine models were employed to evaluate the correlation between GPR109A and liver cancer progression. GPR109A knockout mice, immune cells depletion and primary cell coculture models were used to determine the regulation of GPR109A on tumour microenvironment and identify the underlying mechanism responsible for the formation of intratumour GPR109A
+ myeloid cells., Results: We demonstrate that glutamine shortage in liver cancer tumour microenvironment drives an immunosuppressive GPR109A+ myeloid cells infiltration, leading to the evasion of immune surveillance. Blockade of GPR109A decreases G-MDSCs and M2-like TAMs abundance to trigger the antitumour responses of CD8+ T cells and further improves the immunotherapy efficacy against liver cancer. Mechanistically, tumour cells and tumour-infiltrated myeloid cells compete for glutamine uptake via the transporter SLC1A5 to control antitumour immunity, which disrupts the endoplasmic reticulum (ER) homoeostasis and induces unfolded protein response of myeloid cells to promote GPR109A expression through IRE1α/XBP1 pathway. The restriction of glutamine uptake in liver cancer cells, as well as the blockade of IRE1α/XBP1 signalling or glutamine supplementation, can eliminate the immunosuppressive effects of GPR109A+ myeloid cells and slow down tumour progression., Conclusion: Our findings identify the immunometabolic crosstalk between liver cancer cells and myeloid cells facilitates tumour progression via a glutamine metabolism/ER stress/GPR109A axis, suggesting that GPR109A can be exploited as an immunometabolic checkpoint and putative target for cancer treatment., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2025. No commercial re-use. See rights and permissions. Published by BMJ Group.)- Published
- 2025
- Full Text
- View/download PDF
174. Dual Inhibitors of SARS-CoV-2 3CL Protease and Human Cathepsin L Containing Glutamine Isosteres Are Anti-CoV-2 Agents.
- Author
-
Kumar V, Zhu J, Chenna BC, Hoffpauir ZA, Rademacher A, Rogers AM, Tseng CT, Drelich A, Farzandh S, Lamb AL, and Meek TD
- Subjects
- Humans, Protease Inhibitors pharmacology, Protease Inhibitors chemistry, Protease Inhibitors chemical synthesis, COVID-19 Drug Treatment, Cathepsin L antagonists & inhibitors, Cathepsin L metabolism, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus 3C Proteases metabolism, Coronavirus 3C Proteases chemistry, Glutamine chemistry, Glutamine metabolism, Antiviral Agents pharmacology, Antiviral Agents chemistry, Antiviral Agents chemical synthesis
- Abstract
SARS-CoV-2 3CL protease (Main protease) and human cathepsin L are proteases that play unique roles in the infection of human cells by SARS-CoV-2, the causative agent of COVID-19. Both proteases recognize leucine and other hydrophobic amino acids at the P
2 position of a peptidomimetic inhibitor. At the P1 position, cathepsin L accepts many amino acid side chains, with a partial preference for phenylalanine, while 3CL-PR protease has a stringent specificity for glutamine or glutamine analogues. We have designed, synthesized, and evaluated peptidomimetic aldehyde dual-target (dual-acting) inhibitors using two peptide scaffolds based on those of two Pfizer 3CL-PR inhibitors, Nirmatrelvir , and PF-835321 . Our inhibitors contain glutamine isosteres at the P1 position, including 2-pyridon-3-yl-alanine, 3-pyridinyl-alanine, and 1,3-oxazo-4-yl-alanine groups. Inhibition constants for these new inhibitors ranged from Ki = 0.6-18 nM (cathepsin L) and Ki = 2.6-124 nM (3CL-PR), for which inhibitors with the 2-pyridon-3-yl-alanal substituent were the most potent for 3CL-PR. The anti-CoV-2 activity of these inhibitors ranged from EC50 = 0.47-15 μM. X-ray structures of the peptidomimetic aldehyde inhibitors of 3CL-PR with similar scaffolds all demonstrated the formation of thiohemiacetals with Cys145 , and hydrogen-bonding interactions with the heteroatoms of the pyridon-3-yl-alanyl group, as well as the nitrogen of the N-terminal indole and its appended carbonyl group at the P3 position. The absence of these hydrogen bonds for the inhibitors containing the 3-pyridinyl-alanyl and 1,3-oxazo-4-yl-alanyl groups was reflected in the less potent inhibition of the inhibitors with 3CL-PR. In summary, our studies demonstrate the value of a second generation of cysteine protease inhibitors that comprise a single agent that acts on both human cathepsin L and SARS-CoV-2 3CL protease. Such dual-target inhibitors will provide anti-COVID-19 drugs that remain active despite the development of resistance due to mutation of the viral protease. Such dual-target inhibitors are more likely to remain useful therapeutics despite the emergence of inactivating mutations in the viral protease because the human cathepsin L will not develop resistance. This particular dual-target approach is innovative since one of the targets is viral (3CL-PR) required for viral protein maturation and the other is human (hCatL) which enables viral infection.- Published
- 2025
- Full Text
- View/download PDF
175. Addressing genome scale design tradeoffs in Pseudomonas putida for bioconversion of an aromatic carbon source.
- Author
-
Banerjee D, Menasalvas J, Chen Y, Gin JW, Baidoo EEK, Petzold CJ, Eng T, and Mukhopadhyay A
- Subjects
- Genetic Engineering, Gene Deletion, Proteomics, Citric Acid Cycle genetics, Genome, Bacterial, Pseudomonas putida genetics, Pseudomonas putida growth & development, Pseudomonas putida metabolism, Coumaric Acids metabolism, Glutamine metabolism, Piperidones metabolism, Fumarate Hydratase genetics, Fumarate Hydratase metabolism
- Abstract
Genome-scale metabolic models (GSMM) are commonly used to identify gene deletion sets that result in growth coupling and pairing product formation with substrate utilization and can improve strain performance beyond levels typically accessible using traditional strain engineering approaches. However, sustainable feedstocks pose a challenge due to incomplete high-resolution metabolic data for non-canonical carbon sources required to curate GSMM and identify implementable designs. Here we address a four-gene deletion design in the Pseudomonas putida KT2440 strain for the lignin-derived non-sugar carbon source, p-coumarate (p-CA), that proved challenging to implement. We examine the performance of the fully implemented design for p-coumarate to glutamine, a useful biomanufacturing intermediate. In this study glutamine is then converted to indigoidine, an alternative sustainable pigment and a model heterologous product that is commonly used to colorimetrically quantify glutamine concentration. Through proteomics, promoter-variation, and growth characterization of a fully implemented gene deletion design, we provide evidence that aromatic catabolism in the completed design is rate-limited by fumarase hydratase (FUM) enzyme activity in the citrate cycle and requires careful optimization of another fumarate hydratase protein (PP_0897) expression to achieve growth and production. A double sensitivity analysis also confirmed a strict requirement for fumarate hydratase activity in the strain where all genes in the growth coupling design have been implemented. Metabolic cross-feeding experiments were used to examine the impact of complete removal of the fumarase hydratase reaction and revealed an unanticipated nutrient requirement, suggesting additional functions for this enzyme. While a complete implementation of the design was achieved, this study highlights the challenge of completely inactivating metabolic reactions encoded by under-characterized proteins, especially in the context of multi-gene edits., Competing Interests: Competing interests: The authors declare no competing interests. T.E., D.B., and A.M. have a patent on the topic of bioproduction in Pseudomonas putida (Genetically modified bacterial cells and methods useful for producing indigoidine, US Patent 11,767,521, 2023)., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
176. d-[5- 11 C]-Glutamine Positron Emission Tomography Imaging for Diagnosis and Therapeutic Monitoring of Orthopedic Implant Infections.
- Author
-
Co CM, Mulgaonkar A, Zhou N, Nguyen TP, Harris S, Sherwood A, Ea V, Rubitschung K, Castellino L, Öz OK, Sun X, and Tang L
- Subjects
- Animals, Rats, Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Rats, Sprague-Dawley, Carbon Radioisotopes, Disease Models, Animal, Male, Radiopharmaceuticals chemistry, Fluorodeoxyglucose F18, Positron-Emission Tomography methods, Staphylococcus aureus, Staphylococcal Infections diagnostic imaging, Staphylococcal Infections drug therapy, Staphylococcal Infections microbiology, Prosthesis-Related Infections diagnostic imaging, Prosthesis-Related Infections microbiology, Prosthesis-Related Infections drug therapy, Biofilms drug effects, Glutamine metabolism
- Abstract
Orthopedic implant infections (OIIs) present diagnostic and therapeutic challenges, owing to the lack of methods to distinguish between active infection and sterile inflammation. To address this unmet need, d-amino-acid-based radiotracers with unique metabolic profiles in microorganisms have emerged as a novel class of infection-specific imaging agents. Given the pivotal role of d-glutamine in bacterial biofilm formation and virulence, herein, we explored the potential of positron emission tomography (PET) imaging with d-[5-
11 C]-Glutamine (d-[5-11 C]-Gln) for early detection and treatment monitoring of OIIs. In vitro studies confirmed an active uptake of d-[5-11 C]-Gln by Staphylococcus aureus ( S. aureus ) biofilm commonly associated with OIIs. In vivo evaluations included PET imaging comparisons with d-[5-11 C]-Gln vs l-[5-11 C]-Gln or 2-deoxy-2-[18 F]-fluoroglucose ([18 F]-FDG) in a rat OII model with tibial implantation of sterile or S. aureus - colonized stainless-steel screws before and after treatment. These studies demonstrated that the uptake of d-[5-11 C]-Gln was significantly higher in the infected screws than that in sterile screws (∼3.4-fold, p = 0.008), which displayed significantly higher infection-to-background muscle uptake ratios (∼2-fold, p = 0.011) with d-[5-11 C]-Gln as compared to l-[5-11 C]-Gln. Following a 3 week vancomycin treatment, imaging with d-[5-11 C]-Gln showed a significant reduction in uptake at the infected sites (∼3-fold, p = 0.0008). Further regression analyses revealed a superior correlation of residual infection-associated radiotracer uptake with the postimaging ex vivo bacterial counts for d-[5-11 C]-Gln ( k = 0.473, R2 = 0.796) vs [18 F]-FDG ( k = 0.212, R2 = 0.434), suggesting that d-[5-11 C]-Gln PET had higher sensitivity for detecting residual bacterial burden than [18 F]-FDG PET. Our results demonstrate the translational potential of d-[5-11 C]-Gln PET imaging for noninvasive detection and treatment monitoring of OIIs.- Published
- 2025
- Full Text
- View/download PDF
177. Reproducibility and repeatability of 18F-(2S, 4R)-4-fluoroglutamine PET imaging in preclinical oncology models.
- Author
-
Ayers GD, Cohen AS, Bae SW, Wen X, Pollard A, Sharma S, Claus T, Payne A, Geng L, Zhao P, Tantawy MN, Gammon ST, and Manning HC
- Subjects
- Animals, Mice, Reproducibility of Results, Cell Line, Tumor, Humans, Radiopharmaceuticals, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms pathology, Female, Fluorine Radioisotopes, Positron-Emission Tomography methods, Glutamine metabolism, Glutamine analogs & derivatives
- Abstract
Introduction: Measurement of repeatability and reproducibility (R&R) is necessary to realize the full potential of positron emission tomography (PET). Several studies have evaluated the reproducibility of PET using 18F-FDG, the most common PET tracer used in oncology, but similar studies using other PET tracers are scarce. Even fewer assess agreement and R&R with statistical methods designed explicitly for the task. 18F-(2S, 4R)-4-fluoro-glutamine (18F-Gln) is a PET tracer designed for imaging glutamine uptake and metabolism. This study illustrates high reproducibility and repeatability with 18F-Gln for in vivo research., Methods: Twenty mice bearing colorectal cancer cell line xenografts were injected with ~9 MBq of 18F-Gln and imaged in an Inveon microPET. Three individuals analyzed the tumor uptake of 18F-Gln using the same set of images, the same image analysis software, and the same analysis method. Scans were randomly re-ordered for a second repeatability measurement 6 months later. Statistical analyses were performed using the methods of Bland and Altman (B&A), Gauge Reproducibility and Repeatability (Gauge R&R), and Lin's Concordance Correlation Coefficient. A comprehensive equivalency test, designed to reject a null hypothesis of non-equivalence, was also conducted., Results: In a two-way random effects Gauge R&R model, variance among mice and their measurement variance were 0.5717 and 0.024. Reproducibility and repeatability accounted for 31% and 69% of the total measurement error, respectively. B&A repeatability coefficients for analysts 1, 2, and 3 were 0.16, 0.35, and 0.49. One-half B&A agreement limits between analysts 1 and 2, 1 and 3, and 2 and 3 were 0.27, 0.47, and 0.47, respectively. The mean square deviation and total deviation index were lowest for analysts 1 and 2, while coverage probabilities and coefficients of the individual agreement were highest. Finally, the definitive agreement inference hypothesis test for equivalency demonstrated that all three confidence intervals for the average difference of means from repeated measures lie within our a priori limits of equivalence (i.e. ± 0.5%ID/g)., Conclusions: Our data indicate high individual analyst and laboratory-level reproducibility and repeatability. The assessment of R&R using the appropriate methods is critical and should be adopted by the broader imaging community., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2025 Ayers et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2025
- Full Text
- View/download PDF
178. Gefitinib Reverses PD-L1-Mediated Immunosuppression Induced by Long-term Glutamine Blockade in Bladder Cancer.
- Author
-
Ma G, Jia H, Li Z, Zhang X, Wang L, Zhang Z, Xiao Y, Liang Z, Li D, Chen Y, Tian X, Wang Y, Liang Y, and Niu H
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Diazooxonorleucine pharmacology, Diazooxonorleucine therapeutic use, Female, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Immune Tolerance drug effects, Signal Transduction drug effects, Immunosuppression Therapy, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms immunology, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, Glutamine metabolism, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen metabolism, Gefitinib pharmacology, Gefitinib therapeutic use
- Abstract
Glutamine is a major energy source for tumor cells, and blocking glutamine metabolism is being investigated as a promising strategy for cancer therapy. However, the antitumor effect of glutamine blockade in bladder cancer remains unclear, necessitating further investigation. In this study, we demonstrated that glutamine metabolism was involved in the malignant progression of bladder cancer. Treatment with the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON) inhibited the growth of bladder cancer cells in vitro in several ways. In addition, we observed inhibition of tumor growth in bladder cancer-bearing mice by using JHU083, a prodrug that was designed to prevent DON-induced toxicity. However, the antitumor immune effect of T cells changed from activation to inhibition as the administrated time extended. We found that both in vitro treatment with DON and in vivo prolonged administration of JHU083 led to the upregulation of PD-L1 in bladder cancer cells. Mechanistically, glutamine blockade upregulated PD-L1 expression in bladder cancer cells by accumulating reactive oxygen species, subsequently activating the EGFR/ERK/C-Jun signaling pathway. Combination treatment of JHU083 and gefitinib reversed the upregulation of PD-L1 in bladder cancer cells induced by prolonged glutamine blockade, resulting in the alleviation of T-cell immunosuppression and a significant improvement in therapeutic outcome. These preclinical findings show promise for glutamine metabolism targeting as a viable therapeutic strategy for bladder cancer, with the potential for further enhancement through combined treatment with gefitinib., (©2024 American Association for Cancer Research.)
- Published
- 2025
- Full Text
- View/download PDF
179. Folate-targeted nanoparticles for glutamine metabolism inhibition enhance anti-tumor immunity and suppress tumor growth in ovarian cancer.
- Author
-
Chen S, Jiang Y, Zheng J, Li P, Liu M, Zhu Y, Zhu S, and Chang S
- Abstract
Ovarian cancer (OC) is a highly malignant gynecological tumor, and its effective treatment is frequently impeded by drug resistance and recurrent tumor growth. The reprogramming of glutamine metabolism in ovarian cancer is closely associated with tumor progression and the immunosuppressive tumor microenvironment. Recently, targeting metabolic reprogramming has emerged as a promising approach for cancer therapy. However, the application of such therapies is often constrained by their significant toxicity to normal tissues. In this study, we fabricated folate-targeted nanoparticles (FA-DCNPs) that co-encapsulate the glutamine metabolism inhibitor 6-diazo-5-oxo-L-norleucine (DON) and calcium carbonate (CaCO
3 ). These nanoparticles alleviate damage to normal tissues by specifically targeting tumor cells via folate receptors (FOLR) mediation. Under acidic conditions, the FA-DCNPs release DON and Ca2+ , generating a synergistic anti-tumor effect by impeding glutamine metabolism and inducing calcium overload. Additionally, FA-DCNPs target M2 phenotype tumor-associated macrophages (TAMs) via FOLR2, attenuating M2-TAMs activity. When partially phagocytosed by M0-TAMs, the nanoparticles restrict glutamate production, inhibiting polarization towards the M2 phenotype. This resulted in an increased proportion of M1-TAMs, thereby improving the tumor immune microenvironment. Our study explores a nanotherapeutic strategy that enhances the biosafety of anti-glutamine metabolism therapy through folate targeting, effectively suppresses tumor cell proliferation, and enhances the anti-tumor immune response., Competing Interests: Declaration of competing interest The authors report that they have no known competing financial interests or personal relationships that could have had the appearance of influencing the work reported in this paper., (Copyright © 2025 Elsevier B.V. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
180. Glutaminase-2 Expression Induces Metabolic Changes and Regulates Pyruvate Dehydrogenase Activity in Glioblastoma Cells.
- Author
-
De Los Santos-Jiménez J, Campos-Sandoval JA, Rosales T, Ko B, Alonso FJ, Márquez J, DeBerardinis RJ, and Matés JM
- Subjects
- Humans, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Glutamine metabolism, Benzeneacetamides pharmacology, Citric Acid Cycle, Thiadiazoles pharmacology, Phosphorylation, Brain Neoplasms metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology, Glutaminase metabolism, Glutaminase genetics, Glioblastoma metabolism, Glioblastoma genetics, Glioblastoma pathology
- Abstract
Glutaminase controls the first step in glutaminolysis, impacting bioenergetics, biosynthesis and oxidative stress. Two isoenzymes exist in humans, GLS and GLS2. GLS is considered prooncogenic and overexpressed in many tumours, while GLS2 may act as prooncogenic or as a tumour suppressor. Glioblastoma cells usually lack GLS2 while they express high GLS. We investigated how GLS2 expression modifies the metabolism of glioblastoma cells, looking for changes that may explain GLS2's potential tumour suppressive role. We developed LN-229 glioblastoma cells stably expressing GLS2 and performed isotope tracing using U-
13 C-glutamine and metabolomic quantification to analyze metabolic changes. Treatment with GLS inhibitor CB-839 was also included to concomitantly inhibit endogenous GLS. GLS2 overexpression resulted in extensive metabolic changes, altering the TCA cycle by upregulating part of the cycle but blocking the synthesis of the 6-carbon intermediates from acetyl-CoA. Expression of GLS2 caused downregulation of PDH activity through phosphorylation of S293 of PDHA1. GLS2 also altered nucleotide levels and induced the accumulation of methylated metabolites and S-adenosyl methionine. These changes suggest that GLS2 may be a key regulator linking glutamine and glucose metabolism, also impacting nucleotides and epigenetics. Future research should ascertain the mechanisms involved and the generalizability of these findings in cancer or physiological conditions.- Published
- 2025
- Full Text
- View/download PDF
181. Inhibition of glutaminase 1 reduces M1 macrophage polarization to protect against monocrotaline-induced pulmonary arterial hypertension.
- Author
-
Chen X, Li L, Deng Y, Liao J, Meng H, Liang L, Hu J, Xie D, and Liang G
- Abstract
(1) BACKGROUND: Metabolic abnormalities and immune inflammation are key elements within pathogenesis of pulmonary arterial hypertension (PAH). And in PAH patients, aberrant glutamine metabolism has been observed; however, the function of glutaminase 1 (GLS1) in macrophage is still unknown. So we aims to investigate GLS1's impact upon macrophages in PAH. (2) METHODS: We firstly constructed an monocrotaline (MCT)-induced PAH rat model. Briefly, the PAH rats were treated with the GLS1 inhibitor BPTES, and various index were evaluated, including hemodynamics, right ventricular function, pulmonary vascular remodeling, macrophage markers, and glutamine metabolism. After that, we polarized bone marrow-derived macrophages (BMDMs) into M1 phenotype and then subjected to BPTES intervention. Finally, we assessed macrophage phenotype, inflammatory markers, and glutamine metabolism indicators, along with the impact of BMDM supernatant on the behavior of pulmonary arterial smooth muscle cells (PASMCs). (3) RESULTS: GLS1 was significantly upregulated in both PAH patients and rats. Treatment with the GLS1 inhibitor BPTES markedly improved pulmonary arterial pressure, right ventricular function, and pulmonary vascular remodeling in PAH rats, while inhibiting M1 macrophage polarization, NLRP3 activation, and the release of pro-inflammatory cytokines. This, in turn, alleviated the proliferation and migration of PASMCs induced by inflammatory stimuli. (4) CONCLUSION: We propose that targeting GLS1 to reduce M1 macrophage polarization and inflammatory responses may represent a promising therapeutic approach for PAH., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2025 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
182. Targeting Asparagine Metabolism in Solid Tumors.
- Author
-
Hanada K, Kawada K, and Obama K
- Subjects
- Humans, Antineoplastic Agents therapeutic use, Antineoplastic Agents pharmacology, Animals, Asparagine metabolism, Neoplasms metabolism, Energy Metabolism
- 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.
- Published
- 2025
- Full Text
- View/download PDF
183. Mitochondrial-cytochrome c oxidase II promotes glutaminolysis to sustain tumor cell survival upon glucose deprivation.
- Author
-
Yi Y, Wang G, Zhang W, Yu S, Fei J, An T, Yi J, Li F, Huang T, Yang J, Niu M, Wang Y, Xu C, and Xiao ZJ
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic, Tumor Microenvironment, Flavin-Adenine Dinucleotide metabolism, Signal Transduction, Mice, Nude, Female, Glucose metabolism, Glutamine metabolism, Cell Survival, Glutaminase metabolism, Glutaminase genetics, Electron Transport Complex IV metabolism, Electron Transport Complex IV genetics, Mitochondria metabolism
- Abstract
Glucose deprivation, a hallmark of the tumor microenvironment, compels tumor cells to seek alternative energy sources for survival and growth. Here, we show that glucose deprivation upregulates the expression of mitochondrial-cytochrome c oxidase II (MT-CO2), a subunit essential for the respiratory chain complex IV, in facilitating glutaminolysis and sustaining tumor cell survival. Mechanistically, glucose deprivation activates Ras signaling to enhance MT-CO2 transcription and inhibits IGF2BP3, an RNA-binding protein, to stabilize MT-CO2 mRNA. Elevated MT-CO2 increases flavin adenosine dinucleotide (FAD) levels in activating lysine-specific demethylase 1 (LSD1) to epigenetically upregulate JUN transcription, consequently promoting glutaminase-1 (GLS1) and glutaminolysis for tumor cell survival. Furthermore, MT-CO2 is indispensable for oncogenic Ras-induced glutaminolysis and tumor growth, and elevated expression of MT-CO2 is associated with poor prognosis in lung cancer patients. Together, these findings reveal a role for MT-CO2 in adapting to metabolic stress and highlight MT-CO2 as a putative therapeutic target for Ras-driven cancers., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
184. Asparagine synthetase and G-protein coupled estrogen receptor are critical responders to nutrient supply in KRAS mutant colorectal cancer.
- Author
-
Lu L, Zhang Q, Aladelokun O, Berardi D, Shen X, Marin A, Garcia-Milian R, Roper J, Khan SA, and Johnson CH
- Subjects
- Humans, Female, Cell Line, Tumor, Male, Glutamine metabolism, Nutrients metabolism, Estradiol pharmacology, Estradiol metabolism, Gene Expression Regulation, Neoplastic, Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor, Receptors, G-Protein-Coupled metabolism, Receptors, G-Protein-Coupled genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms metabolism, Receptors, Estrogen metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Cell Proliferation, Aspartate-Ammonia Ligase metabolism, Aspartate-Ammonia Ligase genetics, Mutation
- Abstract
Survival differences exist in colorectal cancer (CRC) patients by sex and disease stage. However, the potential molecular mechanism(s) are not well understood. Here we show that asparagine synthetase (ASNS) and G protein-coupled estrogen receptor-1 (GPER1) are critical sensors of nutrient depletion and linked to poorer outcomes for females with CRC. Using a 3D spheroid model of isogenic SW48 KRAS wild-type (WT) and G12A mutant (MT) cells grown under a restricted nutrient supply, we found that glutamine depletion inhibited cell growth in both cell lines, whereas ASNS and GPER1 expression were upregulated in KRAS MT versus WT. Estradiol decreased growth in KRAS WT but had no effect on MT cells. Selective GPER1 and ASNS inhibitors suppressed cell proliferation with increased caspase-3 activity of MT cells under glutamine depletion condition particularly in the presence of estradiol. In a clinical colon cancer cohort from The Cancer Genome Atlas, both high GPER1 and ASNS expression were associated with poorer overall survival for females only in advanced stage tumors. These results suggest KRAS MT cells have mechanisms in place that respond to decreased nutrient supply, typically observed in advanced tumors, by increasing the expression of ASNS and GPER1 to drive cell growth. Furthermore, KRAS MT cells are resistant to the protective effects of estradiol under nutrient deplete conditions. The findings indicate that GPER1 and ASNS expression, along with the interaction between nutrient supply and KRAS mutations shed additional light on the mechanisms underlying sex differences in metabolism and growth in CRC, and have clinical implications in the precision management of KRAS mutant CRC., (© 2024 UICC.)
- Published
- 2025
- Full Text
- View/download PDF
185. AdipoRon mitigates liver fibrosis by suppressing serine/glycine biosynthesis through ATF4-dependent glutaminolysis.
- Author
-
Zhang X, Zeng Y, Ying H, Hong Y, Xu J, Lin R, Chen Y, Wu X, Cai W, Xia Z, Zhao Q, Wang Y, Zhou R, Zhu D, and Yu F
- Subjects
- Animals, Mice, Male, Endoplasmic Reticulum Stress drug effects, Mice, Inbred C57BL, Glutamine metabolism, Humans, Carbon Tetrachloride toxicity, Collagen biosynthesis, Cell Line, Piperidines, Glycine analogs & derivatives, Glycine pharmacology, Liver Cirrhosis pathology, Liver Cirrhosis chemically induced, Liver Cirrhosis drug therapy, Activating Transcription Factor 4 metabolism, Serine, Hepatic Stellate Cells drug effects, Hepatic Stellate Cells metabolism
- Abstract
AdipoRon has been validated for its ability to reverse liver fibrosis, yet the underlying mechanisms remain to be thoroughly investigated. Collagen, predominantly synthesized and secreted in hepatic stellate cells (HSCs), relies on glycine as a crucial constituent. Activating transcription factor 4 (ATF4) serves as a pivotal transcriptional regulator in amino acid metabolism. Therefore, our objective is to explore the impact of AdipoRon on ATF4-mediated endoplasmic reticulum stress and amino acid metabolism in HSCs. We induced liver fibrosis in mice through intraperitoneal injection of CCl
4 and administered AdipoRon (50 mg/kg) via gavage. In vitro studies were predominantly conducted using LX-2 cells. Our findings demonstrated that AdipoRon effectively suppressed ATF4-mediated endoplasmic reticulum stress in HSCs and assumed a crucial role in hindering serine/glycine biosynthesis. Interestingly, this inhibitory effect of AdipoRon on serine/glycine biosynthesis is regulated by PSAT1-mediated glutaminolysis, resulting in a subsequent decrease in collagen synthesis within HSCs. This study provides potential mechanistic insights into the treatment of liver fibrosis with AdipoRon., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)- Published
- 2025
- Full Text
- View/download PDF
186. Salinity-Induced Photorespiration in Populus Vascular Tissues Facilitate Nitrogen Reallocation.
- Author
-
Wilhelmi MDMR, Maneejantra N, Balasubramanian VK, Purvine SO, Williams S, DiFazio S, Stewart CN Jr, Ahkami AH, and Blumwald E
- Subjects
- Plant Proteins metabolism, Plant Proteins genetics, Plant Leaves metabolism, Plant Leaves physiology, Gene Expression Regulation, Plant drug effects, Plant Roots metabolism, Plant Roots physiology, Plant Vascular Bundle physiology, Plant Vascular Bundle drug effects, Plant Vascular Bundle metabolism, Glutamate-Ammonia Ligase metabolism, Glutamate-Ammonia Ligase genetics, Sodium Chloride pharmacology, Glutamine metabolism, Populus physiology, Populus metabolism, Populus genetics, Populus drug effects, Nitrogen metabolism, Salinity
- Abstract
Adaptation to abiotic stress is critical for the survival of perennial tree species. Salinity affects plant growth and productivity by interfering with major biosynthetic processes. Detrimental effects of salinity may vary between different plant tissues and cell types. However, spatial molecular mechanisms controlling plant responses to salinity stress are not yet thoroughly understood in perennial trees. We used laser capture microdissection in clones of Populus tremula x alba to isolate palisade and vascular cells of intermediary leaf from plants exposed to 150 mM NaCl for 10 days, followed by a recovery period. Cell-specific changes in proteins and metabolites were determined. Salinity induced a vascular-specific accumulation of proteins associated with photorespiration, and the accumulation of serine, 3-phosphoglycerate and NH
4 + suggesting changes in N metabolism. Accumulation of the GLUTAMINE SYNTHETASE 2 protein, and increased GS1.1 gene expression, indicated that NH4 + produced in photorespiration was assimilated to glutamine, the main amino acid translocated in Populus trees. Further analysis of total soluble proteins in stems and roots showed the accumulation of bark storage proteins induced by the salinity treatments. Collectively, our results suggest that the salt-induced photorespiration in vascular cells mediates N-reallocation in Populus, an essential process for the adaptation of trees to adverse conditions., (© 2024 John Wiley & Sons Ltd.)- Published
- 2025
- Full Text
- View/download PDF
187. Glutamine metabolism is essential for coronavirus replication in host cells and in mice.
- Author
-
Greene KS, Choi A, Yang N, Chen M, Li R, Qiu Y, Ezzatpour S, Rojas KS, Shen J, Wilson KF, Katt WP, Aguilar HC, Lukey MJ, Whittaker GR, and Cerione RA
- Subjects
- Animals, Humans, Mice, Vero Cells, Chlorocebus aethiops, Glutamine metabolism, Glutaminase metabolism, Glutaminase genetics, Virus Replication, SARS-CoV-2 metabolism, COVID-19 metabolism, COVID-19 virology, COVID-19 genetics, COVID-19 pathology
- Abstract
Understanding the fundamental biochemical and metabolic requirements for the replication of coronaviruses within infected cells is of notable interest for the development of broad-based therapeutic strategies, given the likelihood of the emergence of new pandemic-potential virus species, as well as future variants of SARS-CoV-2. Here we demonstrate members of the glutaminase family of enzymes (GLS and GLS2), which catalyze the hydrolysis of glutamine to glutamate (i.e., the first step in glutamine metabolism), play key roles in coronavirus replication in host cells. Using a range of human seasonal and zoonotic coronaviruses, we show three examples where GLS expression increases during coronavirus infection of host cells, and another where GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the "glutamine addiction" of virus-infected cells. We demonstrate that genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, a specific GLS inhibitor, block viral replication in epithelial cells. Moreover, treatment of SARS-CoV-2 infected K18-human ACE2 transgenic mice with SU1 resulted in their complete survival compared to untreated control animals, which succumbed within 10 days post-infection. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and mice and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of broad-based anti-viral drug candidates., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
188. METTL16 controls airway inflammations in smoking-induced COPD via regulating glutamine metabolism.
- Author
-
Jia X, Liu S, Sun C, Zhu M, Yuan Q, Wang M, Xu T, Wang Z, Chen Z, Huang M, Ji N, and Zhang M
- Subjects
- Animals, Mice, Humans, Male, Mice, Inbred C57BL, Smoking adverse effects, Inflammation, Lung pathology, Lung metabolism, Lung drug effects, Female, Disease Models, Animal, Pulmonary Disease, Chronic Obstructive chemically induced, Methyltransferases metabolism, Methyltransferases genetics, Glutamine metabolism
- Abstract
The persistent airway inflammation is the main characteristic of chronic obstructive pulmonary disease (COPD), typically caused by an indoor environment pollution cigarette smoke (CS). METTL16 is an m
6 A methyltransferase that has been proven to be closely associated with the occurrence of various diseases. However, its exact role in smoking-induced COPD remains to be investigated. In this study, we found that the level of METTL16 was aberrantly decreased in lung tissues of COPD smokers. Similarly, murine model induced by CS and lung epithelial cell model induced by cigarette smoke extract (CSE) also confirmed this discovery. Moreover, in the Mettl16-deficient (Mettl16+/- ) mice challenged with CS, airway inflammation was aggravated. To identify the potential target genes and regulatory pathways through METTL16, methylated RNA immunoprecipitation sequencing (meRIP-seq), RNA sequencing (RNA-seq) and metabolomic profiling were used. Knockdown of METTL16 significantly reduced the stability of glutamic-oxaloacetic transaminase 2 (GOT2) and downregulated its expression through m6 A modification, while reprogramed glutamine metabolism in lung epithelial cells. Significant reduction in inflammation levels was observed in the 3-month COPD murine model fed a glutamine-supplemented diet. Mechanistically, METTL16 could regulate lung epithelial mitochondrial function by participating in the reprogramming of glutamine metabolism. Our study characterized the role of the METTL16/GOT2/glutamine axis in the occurrence and development of COPD, and emphasized the potential value of METTL16 and glutamine in the therapy of chronic airway inflammation in smoking-induced COPD., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Mingshun Zhang reports financial support was provided by National Nature Science Foundation of ChinA. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
189. TRIM2 promotes metabolic adaptation to glutamine deprivation via enhancement of CPT1A activity.
- Author
-
Liao K, Liu K, Wang Z, Zhao K, and Mei Y
- Subjects
- Humans, Animals, Mice, Apoptosis, Cell Line, Tumor, Mice, Nude, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Adaptation, Physiological genetics, HEK293 Cells, Glutamine metabolism, Carnitine O-Palmitoyltransferase metabolism, Carnitine O-Palmitoyltransferase genetics, Cell Proliferation, Activating Transcription Factor 4 metabolism, Activating Transcription Factor 4 genetics
- Abstract
Cancer cells undergo metabolic adaptation to promote their survival and growth under energy stress conditions, yet the underlying mechanisms remain largely unclear. Here, we report that tripartite motif-containing protein 2 (TRIM2) is upregulated in response to glutamine deprivation by the transcription factor cyclic AMP-dependent transcription factor (ATF4). TRIM2 is shown to specifically interact with carnitine O-palmitoyltransferase 1 (CPT1A), a rate-limiting enzyme of fatty acid oxidation. Via this interaction, TRIM2 enhances the enzymatic activity of CPT1A, thereby regulating intracellular lipid levels and protecting cells from glutamine deprivation-induced apoptosis. Furthermore, TRIM2 is able to promote both in vitro cell proliferation and in vivo xenograft tumor growth via CPT1A. Together, these findings establish TRIM2 as an important regulator of the metabolic adaptation of cancer cells to glutamine deprivation and implicate TRIM2 as a potential therapeutic target for cancer., (© 2024 Federation of European Biochemical Societies.)
- Published
- 2025
- Full Text
- View/download PDF
190. Decomposition of L-glutamine and accumulation of ammonium in cell culture media inhibit infectivity of influenza viruses.
- Author
-
Kegel NB, Kaufmann A, Matrosovich M, Bauer S, and Dorna J
- Subjects
- Humans, Animals, Influenza A virus drug effects, Influenza A virus physiology, Madin Darby Canine Kidney Cells, Antiviral Agents pharmacology, Dogs, Culture Media chemistry, Glutamine metabolism, Glutamine pharmacology, Ammonium Compounds metabolism, Ammonium Compounds pharmacology
- Abstract
Cationic lysosomotropic molecules such as ammonium salts and chloroquine inhibit influenza A virus (IAV) infection in cell culture by counteracting endosomal acidification and hampering viral-endosomal fusion. Here, we studied the effects of storage of L-glutamine-supplemented cell culture media on accumulation of ammonium and inhibition of IAV infection. The storage-related inhibitory effect was observed for DMEM and OptiMEM media but not for RPMI medium, and was more pronounced for IAV with pH-stable hemagglutinin. Our results highlight the importance to consider potential presence of virus-inhibiting lysosomotropic agents and/or their production in culture media with negative effect on influenza virus infection., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
191. Glutamine Supplementation as a Novel Metabolic Therapeutic Strategy for LIG3-Dependent Chronic Intestinal Pseudo-Obstruction.
- Author
-
Diquigiovanni C, Rizzardi N, Cataldi-Stagetti E, Gozzellino L, Isidori F, Valenti F, Orsini A, Astolfi A, Giangregorio T, Pironi L, Boschetti E, Arrigo S, Maresca A, Magnoni P, Costanzini A, Carelli V, Taniguchi-Ikeda M, Fato R, Bergamini C, De Giorgio R, and Bonora E
- Subjects
- Humans, Male, Chronic Disease, Mitochondria metabolism, Mitochondria drug effects, Female, Cells, Cultured, Dietary Supplements, Gene Expression Profiling, Energy Metabolism drug effects, Case-Control Studies, Intestinal Pseudo-Obstruction genetics, Intestinal Pseudo-Obstruction metabolism, Intestinal Pseudo-Obstruction drug therapy, Fibroblasts metabolism, Fibroblasts drug effects, Fibroblasts pathology, Glutamine metabolism, Mutation, Mitophagy drug effects, DNA Ligase ATP genetics, DNA Ligase ATP metabolism
- Abstract
Background & Aims: We recently identified a recessive syndrome due to DNA ligase 3 (LIG3) mutations in patients with chronic intestinal pseudo-obstruction, leukoencephalopathy, and neurogenic bladder. LIG3 mutations affect mitochondrial DNA maintenance, leading to defective energy production. We aimed at identifying altered molecular pathways and developing possible targeted treatments to revert/ameliorate the cellular energy impairment., Methods: Whole transcriptome analysis was performed on patient-derived fibroblasts total RNA and controls. Mitochondrial function, mitophagy, and l-glutamine supplementation effects were analyzed by live cell analysis, immunostaining, and Western blot. Patients were treated with Dipeptiven (Fresenius-Kabi) according to standard protocols. Patients' symptoms were analyzed by the Gastrointestinal Symptom Rating Scale questionnaire., Results: We identified deregulated transcripts in mutant fibroblasts vs controls, including overexpression of genes involved in extracellular matrix development and remodeling and mitochondrial functions. Gut biopsy specimens of LIG3-mutant patients documented collagen and elastic fiber accumulation. Mutant fibroblasts exhibited impaired mitochondrial mitophagy indicative of dysfunctional turnover and altered Ca
2+ homeostasis. Supplementation with l-glutamine (6 mmol/L), previously shown to increase mitochondrial DNA-defective cell survival, improved growth rate and adenosine 5'-triphosphate production in LIG3-mutant fibroblasts. These data led us to provide parenterally a dipeptide containing l-glutamine to 3 siblings carrying biallelic LIG3 mutations. Compared with baseline, gastrointestinal and extra-gastrointestinal symptoms significantly improved after 8 months of treatment., Conclusions: LIG3 deficiency leads to mitochondrial dysfunction. High levels l-glutamine supplementation were beneficial in LIG3-mutant cells and improved symptom severity without noticeable adverse effects. Our results provide a proof of concept to design ad hoc clinical trials with l-glutamine in LIG3-mutant patients., (Copyright © 2025 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
192. Inhibition of Glutamate-to-Glutathione Flux Promotes Tumor Antigen Presentation in Colorectal Cancer Cells.
- Author
-
Yu T, Van der Jeught K, Zhu H, Zhou Z, Sharma S, Liu S, Eyvani H, So KM, Singh N, Wang J, Sandusky GE, Liu Y, Opyrchal M, Cao S, Wan J, Zhang C, and Zhang X
- Subjects
- Animals, Mice, Humans, Cell Line, Tumor, Antigen Presentation immunology, Disease Models, Animal, Glutaminase metabolism, Glutaminase genetics, Tumor Microenvironment immunology, Colorectal Neoplasms metabolism, Colorectal Neoplasms immunology, Colorectal Neoplasms genetics, Glutamic Acid metabolism, Glutathione metabolism
- 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., (© 2024 The Author(s). Advanced Science published by Wiley‐VCH GmbH.)
- Published
- 2025
- Full Text
- View/download PDF
193. Energy metabolism in osteoprogenitors and osteoblasts: Role of the pentose phosphate pathway.
- Author
-
Catheline SE, Smith CO, McArthur M, Yu C, Brookes PS, and Eliseev RA
- Subjects
- Humans, Cell Differentiation, Glutamine metabolism, Citric Acid Cycle, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells cytology, Mitochondria metabolism, Malates metabolism, Cells, Cultured, Pentose Phosphate Pathway, Osteoblasts metabolism, Osteoblasts cytology, Glycolysis, Glucose metabolism, Energy Metabolism
- Abstract
Bioenergetic preferences of osteolineage cells, including osteoprogenitors and osteoblasts (OBs), are a matter of intense debate. Early studies pointed to OB reliance on glucose and aerobic glycolysis while more recent works indicated the importance of glutamine as a mitochondrial fuel. Aiming to clarify this issue, we performed metabolic tracing of
13 C-labeled glucose and glutamine in human osteolineage cells: bone marrow stromal (a.k.a. mesenchymal stem) cells and bone marrow stromal cell-derived OBs. Glucose tracing showed noncanonical direction of glucose metabolism with high labeling of early glycolytic steps and the pentose phosphate pathway (PPP) but very low labeling of late glycolytic steps and the Krebs cycle. Labeling of Krebs cycle and late steps of glycolysis was primarily from glutamine. These data suggest that in osteolineage cells, glucose is metabolized primarily via the PPP while glutamine is metabolized in the mitochondria, also feeding into the late steps of glycolysis likely via the malate-aspartate shuttle. This metabolic setup did not change after induction of differentiation. To evaluate the importance of this setup for osteolineage cells, we used the inhibitors of either PPP or malate-aspartate shuttle and observed a significant reduction in both cell growth and ability to differentiate. In sum, we observed a distinct metabolic wiring in osteolineage cells with high flux of glucose through the PPP and glutamine flux fueling both mitochondria and late steps of glycolysis. This wiring likely reflects their unique capacity to rapidly proliferate and produce extracellular matrix, e.g., after bone fracture., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
194. NUAKs promote mTOR/c-Myc-induced glucose and glutamine reprogramming for cell growth and metastasis in breast cancer cells.
- Author
-
Worral Wilfred Raj AS and Manoharan R
- Subjects
- Humans, Female, Cell Line, Tumor, Animals, Mice, Signal Transduction drug effects, Neoplasm Metastasis, Gene Expression Regulation, Neoplastic drug effects, Glycolysis drug effects, Glutamine metabolism, TOR Serine-Threonine Kinases metabolism, Glucose metabolism, Breast Neoplasms pathology, Breast Neoplasms metabolism, Breast Neoplasms genetics, Cell Proliferation, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins c-myc genetics
- Abstract
Breast cancer progression and metastasis are closely connected to changes in glucose and glutamine metabolism. While Novel (nua) kinase family 1 (NUAK1) and Novel (nua) kinase family 2 (NUAK2), which are two members of the AMPK-related kinases, have been associated with breast tumorigenesis, their role in the metabolic reprogramming that occurs during breast cancer progression remains unclear. Our research uncovers that NUAKs expression is significantly higher in breast cancer tissues and cell lines, and it is positively related to glycolysis, the pentose phosphate pathway (PPP), glutamine metabolism, and a poor prognosis for breast cancer patients. We show that NUAKs significantly increase metabolic reprogramming, including aerobic glycolysis, PPP, and glutamine metabolism in triple negative breast cancer subtypes but only induce aerobic glycolysis and PPP in luminal breast cancer subtypes to meet the anabolic demands of rapidly dividing breast cancer cells. In contrast, the depletion of NUAKs has the opposite effect. Mechanistic insights reveal that NUAKs activate mammalian target of rapamycin (mTOR) signaling, which in turn upregulates the c-Myc transcription factor, a crucial regulator of glucose and glutamine metabolic gene expression. Moreover, we demonstrate that NUAKs enhance mTOR/c-Myc signaling pathways, leading to increased glucose and glutamine reprogramming, which supports rapid cell proliferation and metastatic potential in breast cancer cells. Importantly, pretreating breast cancer cells with mTOR inhibitors blocked the metabolic reprogramming and tumor-promoting effect of NUAK1/2. Therefore, targeting NUAKs may represent a novel therapeutic strategy for the treatment of breast cancer., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
195. PIKFYVE inhibition induces endosome- and lysosome-derived vacuole enlargement via ammonium accumulation.
- Author
-
Uwada J, Nakazawa H, Kiyoi T, Yazawa T, Muramatsu I, and Masuoka T
- Subjects
- Humans, Cell Line, Tumor, Phosphoinositide-3 Kinase Inhibitors pharmacology, Glutamine metabolism, Male, Ammonia metabolism, Ammonia pharmacology, Vacuoles metabolism, Vacuoles drug effects, Lysosomes metabolism, Lysosomes drug effects, Endosomes metabolism, Endosomes drug effects, Phosphatidylinositol 3-Kinases metabolism, Ammonium Compounds metabolism, Ammonium Compounds pharmacology
- Abstract
FYVE-type zinc finger-containing phosphoinositide kinase (PIKFYVE), which is essential for phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2] production, is an important regulator of lysosomal homeostasis. PIKFYVE dysfunction leads to cytoplasmic vacuolization; however, the underlying mechanism remains unknown. In this study, we explored the cause of vacuole enlargement upon PIKFYVE inhibition in DU145 prostate cancer cells. Enlargement of vacuoles upon PIKFYVE inhibition required glutamine and its metabolism by glutaminases. Addition of ammonia, a metabolite of glutamine, was sufficient to enlarge vacuoles via PIKFYVE inhibition. Moreover, PIKFYVE inhibition led to intracellular ammonium accumulation. Endosome-lysosome permeabilization resulted in ammonium leakage from the cells, indicating ammonium accumulation in the endosomes and lysosomes. Ammonium accumulation and vacuole expansion were suppressed by the lysosomal lumen neutralization. It is therefore assumed that PIKFYVE inhibition interferes with the efflux of NH4+, which formed through protonation of NH3 in the lysosomal lumen, leading to osmotic swelling of vacuoles. Notably, glutamine or ammonium is required for PIKFYVE inhibition-induced suppression of lysosomal function and autophagic flux. In conclusion, this study shows that PIKFYVE inhibition disrupts lysosomal homeostasis via ammonium accumulation., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2025. Published by The Company of Biologists.)
- Published
- 2025
- Full Text
- View/download PDF
196. Derivatives of D(-)-glutamine-based MMP-2 inhibitors as an effective remedy for the management of chronic myeloid leukemia-Part-III: Synthesis, biological screening and in silico binding interaction analysis.
- Author
-
Das S, Patel T, Himaja A, Regula S, Banerjee S, De AK, Qureshi IA, Gayen S, Ghosh B, Adhikari N, and Jha T
- Subjects
- Humans, Structure-Activity Relationship, Molecular Structure, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Molecular Docking Simulation, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Matrix Metalloproteinase 2 metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents chemical synthesis, Matrix Metalloproteinase Inhibitors pharmacology, Matrix Metalloproteinase Inhibitors chemistry, Matrix Metalloproteinase Inhibitors chemical synthesis, Glutamine chemistry, Glutamine metabolism, Glutamine pharmacology, Drug Screening Assays, Antitumor
- Abstract
Tyrosine kinase inhibitors (TKIs) have markedly improved the overall survival rate of patients with chronic myeloid leukemia (CML), enabling them to achieve a normal life expectancy. However, toxicity, relapse, and drug resistance continue to pose major challenges in the clinical treatment of CML. The progression of leukemia is directly connected to higher expression levels and enzymatic actions of matrix metalloproteinase-2 (MMP-2). It is also associated with increased expression and enzymatic actions of matrix metalloproteinase-9 (MMP-9). From this perspective, MMP-2 and MMP-9 offers a promising strategy for developing novel therapeutic molecules that could be effective in treating CML. This study is the Part-III of D(-)-glutamine-based MMP-2 inhibitors series for the management of chronic myeloid leukemia. Fourteen newly synthesized p-tosyl-D(-)-glutamine derivatives were examined in cell culture-based antileukemic assays and also evaluated for their ability to inhibit MMPs. The lead compounds 5g and 5j demonstrated the most promising antileukemic potential. Compounds 5g and 5j are safe for normal cells and effectively block gelatinases (MMP-2 and MMP-9). The best active molecule 5g induced significant apoptosis. Compound 5g reduced MMP-2 levels in the K562 cell line. It also had strong antiangiogenic effects in the ACHN cell line. The strongest MMP-2 inhibitor, 5g, had stable binding at the MMP-2 active site, which is linked to its effective inhibition of MMP-2. In conclusion, these p-tosyl-D(-)-glutamine derivatives are promising MMP-2 inhibitors. They have strong anti-CML effects and should be studied more for future CML treatment., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
197. Alcohol-induced gut microbial reorganization and associated overproduction of phenylacetylglutamine promotes cardiovascular disease.
- Author
-
Li Z, Gu M, Zaparte A, Fu X, Mahen K, Mrdjen M, Li XS, Yang Z, Ma J, Thoudam T, Chandler K, Hesler M, Heathers L, Gorse K, Van TT, Wong D, Gibson AM, Wang Z, Taylor CM, Quijada P, Makarewich CA, Hazen SL, Liangpunsakul S, Brown JM, Lefer DJ, Welsh DA, and Sharp TE 3rd
- Subjects
- Animals, Male, Mice, Humans, Mice, Inbred C57BL, Myocytes, Cardiac metabolism, Myocytes, Cardiac drug effects, Ethanol pharmacology, Ethanol metabolism, Oxidative Stress drug effects, Fecal Microbiota Transplantation, Alcoholism metabolism, Disease Models, Animal, Female, Glutamates metabolism, Endothelial Cells metabolism, Endothelial Cells drug effects, Middle Aged, Gastrointestinal Microbiome drug effects, Glutamine metabolism, Glutamine analogs & derivatives, Cardiovascular Diseases metabolism, Cardiovascular Diseases microbiology, Cardiovascular Diseases etiology
- Abstract
The mechanism(s) underlying gut microbial metabolite (GMM) contribution towards alcohol-mediated cardiovascular disease (CVD) is unknown. Herein we observe elevation in circulating phenylacetylglutamine (PAGln), a known CVD-associated GMM, in individuals living with alcohol use disorder. In a male murine binge-on-chronic alcohol model, we confirm gut microbial reorganization, elevation in PAGln levels, and the presence of cardiovascular pathophysiology. Fecal microbiota transplantation from pair-/alcohol-fed mice into naïve male mice demonstrates the transmissibility of PAGln production and the CVD phenotype. Independent of alcohol exposure, pharmacological-mediated increases in PAGln elicits direct cardiac and vascular dysfunction. PAGln induced hypercontractility and altered calcium cycling in isolated cardiomyocytes providing evidence of improper relaxation which corresponds to elevated filling pressures observed in vivo. Furthermore, PAGln directly induces vascular endothelial cell activation through induction of oxidative stress leading to endothelial cell dysfunction. We thus reveal that the alcohol-induced microbial reorganization and resultant GMM elevation, specifically PAGln, directly contributes to CVD., Competing Interests: Competing interests: Z.W. reports being named as co-inventor on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics and therapeutics. Z.W. also reports having received royalty payments for inventions or discoveries related to cardiovascular diagnostics or therapeutics from Cleveland Heart Lab, a fully owned subsidiary of Quest Diagnostics and Procter & Gamble. C.M.T. consults for Abbott Laboratories. S.L.H. reports being named as co-inventor on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics and therapeutics, being a paid consultant for Procter & Gamble and Zehna Therapeutics, having received research funds from Procter & Gamble, Zehna Therapeutics, and Roche Diagnostics, and being eligible to receive royalty payments for inventions or discoveries related to cardiovascular diagnostics or therapeutics from Cleveland HeartLab and Procter & Gamble. All other authors: Z.L., M.G., A.Z., X.F., K.M., M.M., X.S.L., Z.Y., J.M., T.T., K.C., M.H., L.H., K.G., T.T.V., D.W., A.M.G., P.Q., C.A.M., J.M.B., D.J.L., D.A.W., and T.E.S. have no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
198. RUNX2 enhances bladder cancer progression by promoting glutamine metabolism.
- Author
-
Huang Z, Liu B, Li X, Jin C, Hu Q, Zhao Z, Sun Y, and Wang Q
- Abstract
Bladder cancer is a prevalent malignancy within the urinary system. Prior research has suggested that glutamine metabolism plays a crucial role in driving bladder cancer progression. However, the precise molecular mechanism governing glutamine metabolism in bladder cancer is still inadequately understood. The research revealed a significant correlation between high levels of RUNX2 and SLC7A6 and advanced clinical stage, as well as poor prognosis, in bladder cancer patients. Furthermore, manipulating the levels of RUNX2 through overexpression or silencing demonstrated a significant impact on glutamine and bladder cancer progression. Mechanically, RUNX2 regulates the transcription of SLC7A6, resulting in enhanced glutamine metabolism and promoting the progression of bladder cancer. Overall, this research affirms the crucial function of RUNX2 as a key transcription factor to promoting glutamine and cancer development through modulation of SLC7A6. Targeting RUNX2 could represent a promising therapeutic approach for addressing aberrant glutamine metabolism in bladder cancer., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
199. Dietary glutamine supplementation improves both Th1 and Th17 responses via CARD11-mTORC1 pathway in murine model of atopic dermatitis.
- Author
-
Fan J, Wang X, Wang Y, Song J, Chen M, Weng C, Wang L, Chi Z, and Zhang W
- Subjects
- Animals, Humans, Mice, Amino Acid Transport System ASC genetics, Amino Acid Transport System ASC metabolism, Interferon-gamma metabolism, Interleukin-17 metabolism, Mice, Inbred C57BL, Mice, Knockout, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens metabolism, Pyroglyphidae immunology, Signal Transduction, Skin immunology, Skin pathology, Skin metabolism, Skin drug effects, CARD Signaling Adaptor Proteins metabolism, CARD Signaling Adaptor Proteins genetics, Dermatitis, Atopic immunology, Dermatitis, Atopic drug therapy, Dietary Supplements, Disease Models, Animal, Glutamine metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Th1 Cells immunology, Th17 Cells immunology
- Abstract
Glutamine (GLN) is considered an immunomodulatory nutrient, while caspase recruitment domain 11 (CARD11) is a susceptibility locus for atopic dermatitis (AD). T-cell antigen receptor (TCR)-stimulated GLN uptake requires CARD11. However, the specific pathogenesis of AD via GLN uptake remains unclear. This study aimed to elucidate the association between dietary GLN supplementation and the CARD11 pathway in the pathogenesis of AD, focusing on T helper type 1 (Th1) and Th17 cell expression in AD. Herein, wild-type (WT) mice with house dust mite epidermal-sensitized skin exhibited increased expression of interferon-gamma (IFN-gamma) and interleukin (IL)-17, whereas CARD11 deficiency impaired Th1 and Th17 responses at the same site. CARD11 is a key mediator of Th1 and Th17 expression in AD. Additionally, we suppressed mammalian target of rapamycin complex 1 (mTORC1) signaling, downstream of CARD11, to underscore the critical role of CARD11 in mediating Th1 and Th17 expression in AD. Further, dietary supplementation of GLN to CARD11
-/- mice restored Th1 and Th17 responses, whereas inflammatory expression was reduced in WT mice, and p-CARD11 expression and mTORC1 signaling activity were increased in JPM50.6 cells and CARD11-/- mice. Upon inhibiting the GLN transporter, alanine-serine-cysteine transporter carrier 2 (ASCT2), we observed that the Th1 and Th17 response in AD was reduced. Conclusively, ASCT2-mediated GLN uptake improves the expression of Th1 and Th17 cells via CARD11-mTORC1 signaling pathway in AD, suggesting the potential of glutamine supplementation for AD treatment., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
200. Enhancing GFPT1 expression with glutamine protects chondrocytes in osteoarthritis.
- Author
-
Zhang Z, Li X, Guo W, and Huang Z
- Subjects
- Humans, Animals, Mice, Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) metabolism, Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) genetics, Collagen Type II metabolism, Collagen Type II genetics, Cartilage, Articular metabolism, Cartilage, Articular pathology, Cartilage, Articular drug effects, Interleukin-1beta metabolism, Interleukin-1beta genetics, Cells, Cultured, Chondrocytes metabolism, Chondrocytes drug effects, Osteoarthritis metabolism, Osteoarthritis drug therapy, Glutamine metabolism, Matrix Metalloproteinase 13 metabolism, Matrix Metalloproteinase 13 genetics
- Abstract
Objective: Osteoarthritis (OA) is the leading joint disease without currently available disease-modified drugs. The current study aimed to identify potential drug targets that could decelerate the progression of OA., Methods: We employed Mendelian Randomization (MR) and colocalization analysis to identify therapeutic targets linked to 12 OA traits within 2645 targets. Bulk and single-cell RNA-seq analyses of cartilage samples were conducted to pinpoint GFPT1 and determine the specific cell types in which GFPT1 is expressed. Overexpression and knockdown experiments further explored the expression and potential OA-associated functions of GFPT1., Results: GFPT1 has been identified as a cross-OA therapeutic candidate gene by MR analysis. We observed a significant reduction in GFPT1 expression in OA cartilage compared to normal cartilage from public transcriptomic data of both humans and mice. In vitro experiments confirmed these findings at both mRNA and protein levels in OA chondrocytes. IL-1β stimulation leads to downregulation of GFPT1. We confirmed that supplementary glutamine can reverse the suppression of GFPT1 more effectively than glucosamine in the OA in vitro model. GFPT1 upregulation with glutamine, in turn, further increases the expression of COL2A1 and decreases the expression of MMP13., Conclusions: Our findings demonstrate that GFPT1 is downregulated in OA, and overexpressing GFPT1 can restore the anabolic metabolism of cartilage. Compared to glucosamine, enhancing GFPT1 expression with glutamine to influence the hexosamine biosynthetic pathway may offer a more effective therapeutic strategy for OA., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- 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.