bims-glucam Biomed News
on Glutamine cancer metabolism
Issue of 2025–03–09
sixteen papers selected by
Sreeparna Banerjee, Middle East Technical University



  1. Se Pu. 2025 Mar;43(3): 207-219
      Glutamine synthetase (GS), the only enzyme responsible for de novo glutamine synthesis, plays a significant role in cancer progression. As an example of the consequences of GS mutations, the R324C variant causes congenital glutamine deficiency, which results in brain abnormalities and neonatal death. However, the influence of GS-deficient mutations on cancer cells remains relatively unexplored. In this study, we investigated the effects of GS and GS-deficient mutations, including R324C and previously unreported K241R, which serve as models for GS inactivation. This study provided intriguing insights into the intricate relationship between GS mutations and cancer cell metabolism. Our findings strongly support recent studies that suggest GS deletion leads to the suppression of diverse signaling cascades associated with glutamine metabolism under glutamine-stripping conditions. The affected processes include DNA synthesis, the citric acid cycle, and reactive oxygen species (ROS) detoxification. This suppression originates from the inherent inability of cells to autonomously synthesize glutamine under glutamine-depleted conditions. As a key source of reduced nitrogen, glutamine is crucial for the formation of purine and pyrimidine bases, which are essential building blocks for DNA synthesis. Furthermore, the citric acid cycle is inhibited by the absence of negatively charged glutamate within the mitochondrial matrix, particularly when glutamine is scarce. This deficiency decreases the flux of α-ketoglutarate (α-KG), a principal driver of the citric acid cycle. Intermediate metabolites of the citric acid cycle directly or indirectly contribute to the generation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, a core component of redox homeostasis. Using the GS_R324C and GS_K241R mutants, we conducted an integrative transcriptomics and metabolomics analysis. The GS mutants with reduced activity activated multiple amino acid biosynthesis pathways, including arginine-proline, glycine-serine-threonine, and alanine-aspartate-glutamate metabolism. This intriguing behavior led us to hypothesize that despite hindrance of the citric acid cycle, abundant intracellular glutamate is redirected through alternative processes, including transamination. Simultaneously, key metabolic enzymes in the amino acid synthesis pathways, such as glutamic-oxaloacetic transaminase 1 (GOT1), glutamic-pyruvic transaminase 2 (GPT2), pyrroline-5-carboxylate reductase 1 (PYCR1), and phosphoserine aminotransferase 1 (PSAT1), exhibited increased mRNA levels. Additionally, GS deficiency appeared to upregulate the expression of glutamine transporters SLC38A2 and SLC1A5. Thus, restricting extracellular amino acids, such as glutamine, induces a stress response while promoting transcription or translation by a select group of genes, thereby facilitating cellular adaptation. However, similar to GS_WT, both GS_R324C and GS_K241R were modulated by glutamine treatment. Among GS-activity-dependent behaviors, the increased expression of numerous aminoacyl-tRNA synthetases (ARSs), which are critical for aminoacyl-tRNA biosynthesis, remains poorly understood. Most ARS-encoding genes are transcriptionally induced by activating transcription factor 4 (ATF4), the expression of which increases under oxidative stress, endoplasmic reticulum stress, hypoxia, and amino acid limitation. In GS-deficient cells, the increased expression of ATF4 was accompanied by pronounced stress caused by glutamine starvation. Thus, ARS upregulation may predominantly arise from increased ATF4 expression in GS-deficient cells. Additionally, transcriptomic analysis revealed the differential expression of specific genes, regardless of GS activity, suggesting that GS is involved in various processes other than glutamine synthesis, including angiogenesis. Although our omics study was limited to H1299 cells, in subsequent experiments, we validated our findings using additional cell lines, including Hepa1-6 and LN-229. To attain a more comprehensive understanding of the impact of the newly identified GS_K241R mutant, our investigation should be extended to various cell types and mouse models. In summary, we identified and investigated GS-deficient mutations in cancer cells and conducted an integrative transcriptomics-metabolomics analysis with comparisons to wild-type GS. This comprehensive approach provided crucial insights into the intricate pathways modulated by GS activity. Our findings advance the understanding of how GS functions in the context of reprogrammed cellular metabolism, particularly during glutamine deprivation. The altered metabolism triggered by elevated glutamate levels arising from GS mutations highlights the remarkable plasticity of cancer cell metabolism. Notably, considering the increasing research focus on GS as a potential therapeutic target in various cancer types, the findings of this study could provide innovative perspectives for drug development and the formulation of clinical treatment strategies.
    Keywords:  glutamine; glutamine synthetase mutation; lung cancer; metabolomics; transcriptomics
    DOI:  https://doi.org/10.3724/SP.J.1123.2024.04003
  2. Colloids Surf B Biointerfaces. 2025 Feb 27. pii: S0927-7765(25)00107-9. [Epub ahead of print]251 114600
      Single-atom enzymes (SAZ) show great promise in cancer therapy, particularly chemodynamic therapy, due to their high catalytic activity. They can increase reactive oxygen species (ROS) in tumor cells, causing cell damage and death. However, glutathione (GSH) in tumors can neutralize ROS, reducing SAZ effectiveness. Lowering GSH levels can enhance the effectiveness of SAZ in killing tumor cells, and inhibiting its synthesis at the source might be a promising approach. Glutaminase (GLS1) inhibitors like BPTES can reduce GSH by disrupting glutamine metabolism. This study develops a thermosensitive hydrogel with Fe-based SAZ and BPTES. Upon infrared laser irradiation, the hydrogel releases FeSAZ and BPTES into tumor cells. FeSAZ generates ▪OH from H2O2, while BPTES reduces glutathione (GSH) synthesis in tumor cells, weakening their defenses and enhancing the cytotoxic effects of ▪OH. This combined strategy shows strong potential for effective tumor suppression. Our strategy provides new insights into cancer treatments, potentially offering a more effective therapeutic options for patients.
    Keywords:  BPTES; Glutaminase; Hydrogel; Peroxide; Single-atom enzyme
    DOI:  https://doi.org/10.1016/j.colsurfb.2025.114600
  3. J Inherit Metab Dis. 2025 Mar;48(2): e70018
      The dilated cardiomyopathy with ataxia (DCMA) syndrome is a rare mitochondrial disorder caused by mutations in the poorly understood DNAJC19 gene. Cardiac involvement in DCMA ranges from mild conduction abnormalities to early severe myocardial dysfunction. Although evidence suggests that DCMA is linked to abnormalities in mitochondrial function, the molecular underpinnings of this condition are unclear, and there is no way to predict which patients will develop life-threatening disease. To address this, we developed a metabolic flux assay for assessing the metabolic function of mitochondria in fibroblasts derived from DCMA patients. Using this approach, we discovered that DCMA fibroblasts have elevated glutamine uptake, increased glutamate and ammonium secretion, and elevated lactate production. Moreover, we observed that these cellular perturbations were closely correlated with cardiac dysfunction in a blinded cohort of patient cell lines. These findings suggest that glutamine catabolism is abnormal in DCMA and may serve as a predictor of clinical progression.
    Keywords:  3‐methylglutaconic aciduria; DCMA; dilated cardiomyopathy; glutamine; metabolism
    DOI:  https://doi.org/10.1002/jimd.70018
  4. Nat Commun. 2025 Mar 04. 16(1): 2191
      The deregulated activation of the phosphoinositide 3-kinase (PI3K) pathway is a hallmark of aggressive tumors with metabolic plasticity, eliciting their adaptation to the microenvironment and resistance to chemotherapy. A significant gap lies between the biological features of PI3K-driven tumors and the specific targeting of their vulnerabilities. Here, we explore the metabolic liabilities of PI3K-altered T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological cancer with dismal outcomes. We report a metabolic crosstalk linking glutaminolysis and glycolysis driven by PI3K signaling alterations. Pharmaceutical inhibition of mTOR reveals the singular plasticity of PI3K-altered cells toward the mobilization of glutamine as a salvage pathway to ensure their survival. Subsequently, the combination of glutamine degradation and mTOR inhibition demonstrates robust cytotoxicity in PI3K-driven solid and hematological tumors in pre-clinical and clinical settings. We propose a novel therapeutic strategy to circumvent metabolic adaptation and efficiently target PI3K-driven cancer.
    DOI:  https://doi.org/10.1038/s41467-025-57225-7
  5. Mol Cell Neurosci. 2025 Mar 02. pii: S1044-7431(25)00005-3. [Epub ahead of print]133 103995
      Glutathione (GSH) and heat shock protein 25 (HSP25) reciprocally regulate each other, which maintain redox homeostasis. Since P2X7 receptor (P2X7R) regulates GSH biosynthesis and HSP25 induction, the present study was conducted to explore the role of P2X7R in the reciprocal regulation between HSP25 and GSH in response to kainic acid (KA)-induced nitrosative stress and the related signal pathways, which are largely unknown. The present data demonstrate that P2X7R deletion attenuated KA-induced reductions in total GSH level and nuclear factor-erythroid 2-related factor 2 (Nrf2) intensity/nuclear translocation in astrocytes. P2X7R ablation increased Nrf2 intensity/nuclear translocation in microglia following KA treatment. P2X7R deletion also ameliorated KA-induced inducible nitric oxide synthase (iNOS) and S-nitrosylated-cysteine (SNO-Cys) inductions in microglia and astrocytes. However, P2X7R ablation could not affect KA-induced nuclear Nrf2 translocation and SNO-Cys production in CA3 neurons. Furthermore, P2X7R ablation mitigated S-nitrosylations of glutamine synthase (GS) and alanine-serine-cysteine transporter 2 (ASCT2) induced by KA. HSP25 knockdown increased GSH consumption, astroglial iNOS level and S-nitrosylations of GS and ASCT2, but decreased Nrf2 intensity/nuclear translocation in astrocytes of P2X7R-/- mice following KA injection. These findings indicate that P2X7R facilitated iNOS upregulation by inhibiting HSP25 induction and nuclear Nrf2 translocation in astrocytes, which augmented nitrosative stress-mediated reduction in GSH biosynthesis in response to KA. Therefore, our data suggest that the targeting of P2X7R-Nrf2-iNOS-GS-HSP25 pathway may be required for the maintenance of GSH-mediated redox homeostasis against nitrosative stress, which would prevent the progression of undesirable consequences from seizures and neuroinflammation.
    Keywords:  ASCT2; Glutamate cysteine ligase; Glutamate-glutamine cycle; Glutathione synthetase; S-nitrosylation; Seizure; xCT
    DOI:  https://doi.org/10.1016/j.mcn.2025.103995
  6. Cancer Drug Resist. 2025 ;8 7
      Metabolic reprogramming within the tumor microenvironment (TME) plays a critical role in driving drug resistance in gastrointestinal cancers (GI), particularly through the pathways of fatty acid oxidation and glycolysis. Cancer cells often rewire their metabolism to sustain growth and reshape the TME, creating conditions such as nutrient depletion, hypoxia, and acidity that impair antitumor immune responses. Immune cells within the TME also undergo metabolic alterations, frequently adopting immunosuppressive phenotypes that promote tumor progression and reduce the efficacy of therapies. The competition for essential nutrients, particularly glucose, between cancer and immune cells compromises the antitumor functions of effector immune cells, such as T cells. Additionally, metabolic by-products like lactate and kynurenine further suppress immune activity and promote immunosuppressive populations, including regulatory T cells and M2 macrophages. Targeting metabolic pathways such as fatty acid oxidation and glycolysis presents new opportunities to overcome drug resistance and improve therapeutic outcomes in GI cancers. Modulating these key pathways has the potential to reinvigorate exhausted immune cells, shift immunosuppressive cells toward antitumor phenotypes, and enhance the effectiveness of immunotherapies and other treatments. Future strategies will require continued research into TME metabolism, the development of novel metabolic inhibitors, and clinical trials evaluating combination therapies. Identifying and validating metabolic biomarkers will also be crucial for patient stratification and treatment monitoring. Insights into metabolic reprogramming in GI cancers may have broader implications across multiple cancer types, offering new avenues for improving cancer treatment.
    Keywords:  Gastrointestinal cancers; drug resistance; immune cells; metabolic pathways; tumor microenvironment
    DOI:  https://doi.org/10.20517/cdr.2024.164
  7. Annu Int Conf IEEE Eng Med Biol Soc. 2024 Jul;2024 1-4
      It has been observed that breast cancer is associated with dysregulation of several metabolic pathways that produce metabolite addiction, such as the dependence on glutamine for tumor development. These discoveries might be applied to personalized treatment of this heterogeneous illness by employing specific gene expression or metabolites in cancer therapy. BCAT1 and BCAT2 encode the human branched-chain aminotransferase proteins (hBCAT) involved in cellular metabolism process. Here BCAT2 is exploited through weighted feature-based approach to identify and rank associated genes across public datasets of breast cancer invasive ductal carcinoma patients. BCAT2 lower expression was observed to have the worst prognosis, and BCAT2 gene expression which might be associated with triggering the risk, are ranked, and visualized in different subtypes of breast cancer. These findings give a strong clue to further investigate through experimental approach.
    DOI:  https://doi.org/10.1109/EMBC53108.2024.10782766
  8. MedComm (2020). 2025 Mar;6(3): e70055
      The rising trend in global cancer incidence has caused widespread concern, one of the main reasons being the aging of the global population. Statistical data show that cancer incidence and mortality rates show a clear upward trend with age. Although there is a commonality between dysregulated nutrient sensing, which is one of the main features of aging, and metabolic reprogramming of tumor cells, the specific regulatory relationship is not clear. This manuscript intends to comprehensively analyze the relationship between senescence and tumor metabolic reprogramming; as well as reveal the impact of key factors leading to cellular senescence on tumorigenesis. In addition, this review summarizes the current intervention strategies targeting nutrient sensing pathways, as well as the clinical cases of treating tumors targeting the characteristics of senescence with the existing nanodelivery research strategies. Finally, it also suggests sensible dietary habits for those who wish to combat aging. In conclusion, this review attempts to sort out the link between aging and metabolism and provide new ideas for cancer treatment.
    Keywords:  aging; metabolic reprogramming; mitochondrial dysfunction; signaling pathways; tumor
    DOI:  https://doi.org/10.1002/mco2.70055
  9. Front Immunol. 2025 ;16 1494788
      Metabolic reprogramming is one of the major biological features of malignant tumors, playing a crucial role in the initiation and progression of cancer. The tumor microenvironment consists of various non-cancer cells, such as hepatic stellate cells, cancer-associated fibroblasts (CAFs), immune cells, as well as extracellular matrix and soluble substances. In liver cancer, metabolic reprogramming not only affects its own growth and survival but also interacts with other non-cancer cells by influencing the expression and release of metabolites and cytokines (such as lactate, PGE2, arginine). This interaction leads to acidification of the microenvironment and restricts the uptake of nutrients by other non-cancer cells, resulting in metabolic competition and symbiosis. At the same time, metabolic reprogramming in neighboring cells during proliferation and differentiation processes also impacts tumor immunity. This article provides a comprehensive overview of the metabolic crosstalk between liver cancer cells and their tumor microenvironment, deepening our understanding of relevant findings and pathways. This contributes to further understanding the regulation of cancer development and immune evasion mechanisms while providing assistance in advancing personalized therapies targeting metabolic pathways for anti-cancer treatment.
    Keywords:  hepatocellular carcinoma; immune evasion; metabolism reprogram; signaling pathways; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1494788
  10. Immunology. 2025 Mar 04.
      Cancer immunotherapy has revolutionized treatment paradigms, but its efficacy is often curtailed by T-cell exhaustion and the suppressive tumour microenvironment. Recent studies reveal a novel mechanism of T-cell demise termed ammonia-induced cell death (AICD), which significantly impacts effector CD8+ T-cell survival and function. This phenomenon arises from metabolic reprogramming during immune activation, wherein heightened glutamine metabolism leads to the accumulation of toxic ammonia levels. Ammonia damages lysosomes and mitochondria, disrupting cell balance and causing apoptosis. These insights provide a unique metabolic perspective on T-cell attrition, underscoring the critical interplay between metabolic byproducts and immune regulation. Targeting AICD offers promising therapeutic avenues to bolster immunotherapy. Strategies such as inhibiting ammonia transport, enhancing autophagic pathways and employing ammonia scavengers may extend T-cell longevity and improve antitumor efficacy. Moreover, integrating ammonia modulation with established immunotherapies, including immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, could yield synergistic benefits. Addressing this metabolic bottleneck is particularly compelling in immune 'cold' tumours resistant to conventional therapies. However, further research is essential to refine these interventions, evaluate safety profiles and explore broader applications across cancer types. Ammonia metabolism thus represents a transformative frontier in advancing cancer immunotherapy and precision oncology.
    Keywords:  CD8+ T‐cell; ammonia toxicity; cancer immunotherapy; cell death; tumour
    DOI:  https://doi.org/10.1111/imm.13918
  11. Cell Metab. 2025 Feb 28. pii: S1550-4131(25)00021-X. [Epub ahead of print]
      BRAF V600E-inhibition effectively treats melanoma, but acquired resistance rapidly develops. Protein expression profiles, mitochondrial energetics, metabolomics and fluxomics data in cell line, xenograft, and patient-derived xenograft systems revealed that concerted reprogramming of metabolic pathways (including glutaminolysis, glycolysis, TCA cycle, electron transport chain [ETC], and transsulfuration), along with an immediate cytoprotective response to drug-induced oxidative stress, underpins drug-tolerant persister cancer cell survival. Realignment of cysteine (Cys) metabolism, in particular an immediate upregulation of cystathionine-γ-lyase (CSE), was vital in persister cells. The oxidative cellular environment, drug-induced elevated cystine uptake and oxidative Cys catabolism, increased intracellular cystine/Cys ratios, thereby favoring cystine as a CSE substrate. This produces persulfides and hydrogen sulfide to protect protein thiols and support elevated energy demand in persister cells. Combining BRAF V600E inhibitors with CSE inhibitors effectively diminished proliferative relapse in culture models and increased progression-free survival of xenografted mice. This, together with induced CSE expression in patient samples under BRAF-V600E-inhibition, reveals an approach to increase BRAF-V600E-targeted therapeutic efficacy.
    Keywords:  BRAF V600E targeted therapy resistance; cystathionine γ-lyase; cysteine metabolism; fluxomics of metabolic reprogramming; hydrogen sulfide; melanoma; persister cells; persulfide; redox regulation; transsulfuration
    DOI:  https://doi.org/10.1016/j.cmet.2025.01.021
  12. Hepatol Commun. 2025 Mar 01. pii: e0640. [Epub ahead of print]9(3):
       BACKGROUND: Glutamine (Gln) is a critical amino acid for energy expenditure. It participates in extracellular matrix (ECM) formation and circulates in the hepatic parenchyma in a spatial-oriented manner. Posthepatectomy liver mass recovery poses a regenerative challenge. However, little is known about the role of Gln in liver regeneration, notably the spatial orientation in the remodeling process. This study aimed to elucidate Gln-potentiated liver regeneration and ECM remodeling after mass loss.
    METHODS: We studied the regenerative process in hepatectomized mice supplemented with Gln. Second harmonic generation/two-photon excitation fluorescence microscopy, an artificial intelligence-assisted structure-based imaging, was used to demonstrate the spatial-oriented process in a hepatic acinus.
    RESULTS: Gln promotes liver mass regrowth through the cell cycle, Gln metabolism, and adipogenesis pathways after hepatectomy. Ornithine transaminase, one of the upregulated enzymes, showed temporal, spatial, and functional correspondence with the regeneration process. Second harmonic generation/two-photon excitation fluorescence microscopy highlighted transient hepatic steatosis and ECM collagen synthesis, predominantly in the portal tract instead of the central vein area. Structural remodeling was also observed in the portal tract area.
    CONCLUSIONS: Gln promotes liver regeneration through cellular proliferation and metabolic reprogramming after hepatectomy. Using structure-based imaging, we found that Gln potentiated hepatic steatosis and ECM collagen deposition predominantly in the portal tract area. These results highlighted the spatial orientation and mechanistic implications of Gln in liver regeneration.
    DOI:  https://doi.org/10.1097/HC9.0000000000000640
  13. Cancer Sci. 2025 Mar 05.
      γ-Glutamine cyclotransferase (GGCT) is an enzyme involved in the metabolic cycle of glutathione (GSH). Abnormal GSH metabolism is mostly related to ferroptosis. However, the mechanisms supporting aberrant GGCT expression in PTC remain to be investigated. In this study, we found that GGCT knockdown inhibited GSH synthesis and promoted malondialdehyde (MDA) and reactive oxygen species (ROS) accumulation, thereby promoting ferroptosis in papillary thyroid cancer cells. Pro-GA, the specific inhibitor of GGCT, inhibited the subcutaneous tumor formation of K1 cells. IP combined with LC-MS/MS showed an interaction between GGCT and RPS15A. RPS15A is highly expressed in PTC tissues and cells, and GGCT promotes the stability of RPS15A. Knockdown of RPS15A promoted p53 expression, which in turn inhibited SLC7A11 expression, resulting in ferroptosis, while overexpression of RPS15A reversed GGCT-induced ferroptosis. In addition, miR-205-5p targeted the 3' UTR of GGCT to inhibit GGCT-mediated ferroptosis, tumor growth, and lung metastasis. In conclusion, we found that knockdown of GGCT promoted ferroptosis in PTC cells. Mechanistically, GGCT interacts with RPS15A, and GGCT promotes the protein stability of RPS15A. Knockdown of RPS15A promotes p53 expression and inhibits SLC7A11 expression, thereby inhibiting GSH synthesis. The upstream mechanism of GGCT regulation showed that miR-205-5p inhibited GGCT protein expression by targeting the 3' UTR of GGCT.
    Keywords:  GGCT; RPS15A; ferroptosis; miR‐205‐5p; papillary thyroid carcinoma
    DOI:  https://doi.org/10.1111/cas.70039
  14. J Mol Cell Biol. 2025 Mar 06. pii: mjaf007. [Epub ahead of print]
      Ammonia, traditionally recognized as a toxic nitrogen waste product, has recently emerged as a significant player in diverse physiological processes and implicated in cancer biology. This review article provides an overview of the multifaceted impact of ammonia on cellular signaling pathways, energy metabolism, and tumor microenvironment dynamics, in particular its novel roles in neurotransmission, metabolic homeostasis, cancer cell proliferation, and immune modulation. Notably, ammonia accumulates within the tumor microenvironment, promoting non-essential amino acid synthesis, stimulating mTORC1 activation, promoting lipid synthesis, and impairing various immune cell functions, thereby promoting tumor progression. Furthermore, the potential dual role of ammonia as tumorigenic factor and cancer therapeutic target are discussed, shedding light on its complex regulatory mechanisms and clinical implications. This timely review aims to deepen our understanding of the emerging physiological and pathological roles of ammonia, offering valuable insights into its significance as a potential target for diagnostic and therapeutic interventions in cancer and beyond.
    Keywords:  ammonia; glutamine synthetase; mTOR; tumour microenvironment; urea cycle
    DOI:  https://doi.org/10.1093/jmcb/mjaf007
  15. Cell Rep. 2025 Mar 06. pii: S2211-1247(25)00146-9. [Epub ahead of print]44(3): 115375
      Metabolic enzymes play a central role in cancer metabolic reprogramming, and their dysregulation creates vulnerabilities that can be exploited for therapy. However, accurately measuring metabolic enzyme activity in a high-throughput manner remains challenging due to the complex, multi-layered regulatory mechanisms involved. Here, we present iMetAct, a framework that integrates metabolic-transcription networks with an information propagation strategy to infer enzyme activity from gene expression data. iMetAct outperforms expression-based methods in predicting metabolite conversion rates by accounting for the effects of post-translational modifications. With iMetAct, we identify clinically significant subtypes of hepatocellular carcinoma with distinct metabolic preferences driven by dysregulated enzymes and metabolic regulators acting at both the transcriptional and non-transcriptional levels. Moreover, applying iMetAct to single-cell RNA sequencing data allows for the exploration of cancer cell metabolism and its interplay with immune regulation in the tumor microenvironment. An accompanying online platform further facilitates tumor metabolic analysis, patient stratification, and immune microenvironment characterization.
    Keywords:  CP: Cancer; CP: Metabolism; hepatocellular carcinoma; information propagation; metabolic enzyme activity; tumor stratification; tumor-immune microenvironment
    DOI:  https://doi.org/10.1016/j.celrep.2025.115375
  16. Redox Biol. 2025 Feb 25. pii: S2213-2317(25)00080-1. [Epub ahead of print]81 103567
      Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
    Keywords:  AMPK; Albumin-bound paclitaxel; LKB1; Non-small lung cancer; ROS; SLC7A11
    DOI:  https://doi.org/10.1016/j.redox.2025.103567