bims-meluca Biomed News
on Metabolism of non-small cell lung carcinoma
Issue of 2025–04–13
fourteen papers selected by
the Muñoz-Pinedo/Nadal (PReTT) lab, L’Institut d’Investigació Biomèdica de Bellvitge



  1. Cureus. 2025 Mar;17(3): e80200
       BACKGROUND: Angiopoietin-2 (ANGPT2) and cell migration-inducing protein (CEMIP) are key regulators of angiogenesis, extracellular matrix remodeling, and metastatic progression in various cancers, including lung cancer (LC). The presence of these biomarkers in extracellular vesicles (EVs) may offer valuable insights into the molecular mechanisms underlying LC progression and metastasis. Extracellular vesicles play a critical role in LC by enhancing intercellular communication and supporting processes such as angiogenesis, immune evasion, and metastasis, transferring key molecules like vascular endothelial growth factor (VEGF) and pro-angiogenic microRNAs (miRNAs).
    METHODS: This study aimed to investigate the presence and quantification of ANGPT2 and CEMIP in the cargo of EVs isolated from plasma samples obtained from the peripheral venous blood of patients with localized lung cancer (LLC group), lung cancer with brain metastases (LCM group), and healthy controls (HC group). EVs were isolated using the density gradient ultracentrifugation method, and their characterization was performed through scanning and transmission electron microscopy as well as flow cytometry. Western blot analysis was used to identify ANGPT2 and CEMIP in EV cargo, and band intensity from western blot images was quantified using specialized software.
    RESULTS: The expression of ANGPT2 and CEMIP in EV cargo was significantly higher in the oncologic groups (LLC and LCM combined) compared to the HC group. Notably, EV CEMIP levels were, on average, 59% higher in patients with brain metastases than in those with localized lung cancer. Following surgical resection, postoperative EV ANGPT2 and EV CEMIP levels decreased by 36% and 8.5%, respectively, in the LLC group, and by 40% and 4.6%, respectively, in the LCM group.
    CONCLUSION: These findings emphasize the potential of ANGPT2 and CEMIP as biomarkers for LC progression and prognosis. To our knowledge, no previous study has evaluated the presence and quantification of ANGPT2 and CEMIP in EV cargo from lung cancer patients. To further validate their role in cancer progression, functional studies should explore the mechanistic effects of EV-associated ANGPT2 and CEMIP on angiogenesis, immune modulation, cell migration, extracellular matrix remodeling, and tumor progression in lung cancer models.
    Keywords:  angiopoietin-2; brain metastasis; cell migration-inducing protein; extracellular vesicle; lung cancer
    DOI:  https://doi.org/10.7759/cureus.80200
  2. Cancer Med. 2025 Apr;14(7): e70826
       BACKGROUND: Lung cancer treatment has rapidly advanced, particularly with immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1. However, there are variable responses, such as immune-related adverse events. Several factors predicting the prognosis of lung cancer ICI treatment have been studied so far, but they have limitations, leaving an unmet need. This study aims to investigate delta-He, a novel marker reflecting the iron availability and inflammation through the difference in hemoglobin content between reticulocytes and erythrocytes, as a potential prognostic factor in patients with NSCLC undergoing PD-1/PD-L1 inhibitor therapy.
    METHODS: This research was conducted at Chungnam National University Hospital, analyzing 79 advanced NSCLC patients treated with PD-1/PD-L1 inhibitors. The study population had a mean age of 70 years, with the majority being male (82.3%) and former or current smokers (84.8%). Blood samples collected before therapy initiation were examined for hematological parameters, including delta-He, using Sysmex XN-550 and XN-20 analyzers. The study employed receiver operating characteristic (ROC) curves and Kaplan-Meier curves for the statistical analysis, using SPSS version 26 (IBM Corp., USA) and MedCalc version 22 (MedCalc Software Ltd., Belgium) to evaluate the sensitivity and specificity of delta-He, and to analyze progression-free survival (PFS) and overall survival (OS).
    RESULTS: The study revealed that delta-He is a significant prognostic marker in patients with NSCLC treated with PD-1/PD-L1 inhibitors. A delta-He cutoff value of 3.3 pg was identified based on ROC analysis. Patients with high delta-He values (> 3.3 pg) showed significantly longer median PFS (9.6 vs. 3.0 months, p = 0.024) and OS (not reached vs. 12.2 months, p = 0.010) than those with low values. The high delta-He group also had higher objective response rate (41.4% vs. 26.0%) and disease control rate (86.2% vs. 52.0%). Multivariate analysis highlighted higher delta-He (> 3.3 pg), along with other factors such as FEV1 and smoking status, as important predictors of survival, underscoring its potential role in guiding therapeutic decisions of ICIs in NSCLC (PFS: HR 0.874, 95% CI 0.264-0.874, p = 0.016; OS: HR 0.327, 95% CI 0.150-0.715, p = 0.005).
    CONCLUSION: Our study shows delta-He as a promising prognostic biomarker for lung cancer patients treated with PD-1/PD-L1 inhibitors, highlighting its potential to guide therapeutic decisions and improve patient management in a non-invasive manner. Further research is necessary to validate delta-He's predictive and prognostic value across broader populations and in combination with other biomarkers, emphasizing its role in advancing personalized oncology.
    Keywords:  Delta‐he; NSCLC; PD‐1/PD‐L1 inhibitors; immune checkpoint inhibitors
    DOI:  https://doi.org/10.1002/cam4.70826
  3. Cancer Res. 2025 Apr 08.
      Concurrent inactivating mutations in STK11 and KEAP1 drive primary resistance to therapies, lead to worse outcomes in KRAS-mutated lung adenocarcinoma (KRASmut-LUAD) and are associated with metabolic alterations. Elucidation of the underlying biology of this aggressive LUAD subset is needed to develop effective treatments to improve patient outcomes. Our transcriptomic analysis of 5498 "real-world" KRASmut-LUADs demonstrated that STK11/KEAP1 co-mutation led to upregulation of fatty acid and redox signaling pathways and considerable enrichment of the metabolic genes SCD1 and SLC7A11. High expression of SCD1 and SLC7A11 predicted poor prognosis in KRASmut-patients. Transcriptomics, lipidomics, and kinase arrays in preclinical models demonstrated that SCD1 inhibition promoted ferroptosis, altered fatty acid metabolism, and downregulated SLC7A11 via AKT-GSK3β-NRF2 signaling. SCD1 inhibition caused appreciable tumor regression in xenografts and augmented the efficacy of the ferroptosis inducer erastin. Overall, this study provides insights into the role of the SCD1-SLC7A11 axis in regulating metabolic programming and predicting poor patient outcomes in a genetically defined subset of KRASmut-LUAD.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-24-2745
  4. Mol Genet Genomics. 2025 Apr 08. 300(1): 41
      PPAR γ, as a widely present receptor in tissues, plays a key role in lipid metabolism, energy balance, inflammatory response, and cell differentiation. It plays an important role in the occurrence and development of various tumors, including prostate cancer, gastric cancer, lung cancer, etc., by regulating lipid metabolism. However, the specific mechanism by which it affects lung cancer growth is not yet clear. To investigate how PPAR γ affects lung cancer cell growth by altering ALDH1A3 levels through its impact on lipid metabolism. Bioinformatics analysis was used to predict the correlation between PPAR γ, ALDH1A3 and lung cancer. Based on the results of bioinformatics analysis, PPAR γ activator (Pioglitazone, Pio) and ALDH1A3 inhibitor (diethylaminobenzaldehyde, DEAB) were used to act on lung cancer cells and observe their growth. After measuring the IC50 value of the drug in vitro experiments, lipid metabolomics analysis was conducted to identify the significant changes in differential metabolites and metabolic pathways under the combined influence of Pio and DEAB. Through bioinformatics analysis, it was found that there were significant differences in the levels of PPAR γ and ALDH1A3 between lung cancer and normal lung tissues, and ALDH1A3 was positively correlated with PPAR γ. AUC analysis found that PPAR γ and ALDH1A3 have good predictive value in the diagnosis and prognosis of lung cancer. GSEA enrichment analysis showed that PPAR γ and ALDH1A3 were significantly correlated with lipid oxidation. Combining relevant literature to demonstrate the inhibitory effect of PPAR γ receptors on lung cancer cells and the ability of PPAR γ activation to inhibit ALDH1A3 levels. Further in vitro CCK-8 and IC50 measurements of lung cancer cells A549 and H1299 were conducted, followed by non targeted lipidomics analysis. It was found that the metabolic pathways upregulated by activation of PPAR γ and inhibition of ALDH1A3 included glycerophospholipid metabolism, cholesterol metabolism, arachidonic acid metabolism, and fat digestion and absorption, with glycerophospholipid metabolism pathway accounting for the highest percentage. Conclusion: PPAR γ activation can inhibit the production of ALDH1A3, alter the glycerophospholipid metabolism pathway, and thus inhibit the proliferation of lung cancer cells. This study confirms that PPAR γ affects lung cancer proliferation by influencing the glycerophospholipid metabolism pathway.
    Keywords:  ALDH1A3; Introduce; Lipid metabolism; Lung cancer; PPARγ
    DOI:  https://doi.org/10.1007/s00438-025-02243-9
  5. Int J Endocrinol. 2025 ;2025 7734237
      Objective: This research focused on exploring the shared pathophysiological bases of lung adenocarcinoma (LUAD) and Type 2 diabetes mellitus (T2DM). Methods: The investigation into the molecular similarities between LUAD and T2DM involved querying the Gene Expression Omnibus for pertinent data. Upon pinpointing genes exhibiting differential expression, pathway enrichment analyses were executed to discern the molecular pathways shared by both conditions. In addition, GeneMANIA was employed to establish a protein interaction network, pinpointing STK26 as a critical gene. In addition, the influence of STK26 on the immune environment of the tumor was examined using tools such as the Microenvironment Cell Populations-counter to assess levels of stromal and immune cells in cancer tissues from expression profiles. Furthermore, a lung cancer cell model enriched in glucose was developed to facilitate the knockdown of STK26 using small interfering RNA. The influence of STK26 on A549 cell functionality was assessed using CCK-8, wound healing (scratch), and colony formation (cloning) assays. Results: This will help ensure accuracy and relevance in the revised version. TGF-β, HIF-1, AGE-RAGE, extracellular matrix (ECM) components and function regulation, and cell adhesion were activated in LUAD and T2DM. WGCNA identified two main modules in LUAD, three main modules in T2DM, and 44 shared genes. ClueGO and GeneMANIA analyses focused on pathways regulating cell growth and mitosis. Our analysis revealed STK26 as a central gene that exhibits elevated expression levels in tissues affected by LUAD. Elevated expression of STK26 correlates with a diminished prognosis for LUAD patients. In patients with LUAD characterized by elevated STK26 levels, gene set enrichment analysis identified a notable upregulation in numerous metabolic pathways. These include glycolysis-gluconeogenesis, oxidative phosphorylation, and the conversion pathways between pentose and glucuronic acid, as well as the pentose phosphate pathway. Gene set variation analysis suggested that a high STK26 expression was related to glycolysis, hypoxia, MYC, oxidative phosphorylation, cell cycle, and citric acid cycle pathways. In the group exhibiting elevated levels of STK26, a marked upregulation of glycolytic pathway genes, including HK2, RPIA, IDH3G, and SORD, was noted. This upregulation indicates a correlation between STK26 expression and these pivotal glycolytic genes. MCP-counter analysis suggested that the group with a high STK26 expression level had reduced immune infiltration. Laboratory studies have demonstrated that LUAD cells thrive in a high-glucose setting, where STK26 expression notably surpasses that observed under standard conditions. In addition, suppressing STK26 using siRNA significantly curtails both the growth and movement of LUAD cells. Conclusion: The research established a shared pathogenic basis between LUAD and T2DM. TGF-β, HIF-1, AGE-RAGE, ECM components and function regulation, cell adhesion, and additional signaling pathways are intricately linked with the pathophysiological mechanisms underlying both LUAD and T2DM. Thus, STK26 may affect the development of LUAD and T2DM by regulating glucose metabolism. Suppressing STK26 in a glucose-rich setting curtailed both the expansion and mobility of LUAD cells.
    Keywords:  Alzheimer's disease; LUAD; Type 2 diabetes mellitus; WGCNA; functional enrichment analysis; hub gene; protein–protein interaction network
    DOI:  https://doi.org/10.1155/ije/7734237
  6. Discov Oncol. 2025 Apr 11. 16(1): 515
       BACKGROUND: Lung Adenocarcinoma (LUAD) is a major subtype of Non-Small Cell Lung Cancer (NSCLC) with poor prognosis. Despite advances in molecular targeted therapy and immunotherapy, the five-year survival rate remains low. Disulfidptosis, a novel cell death mechanism, may play a role in tumor progression. CD2AP (CD2-associated protein), a key gene related to Disulfidptosis, is involved in cytoskeleton reorganization and signaling. This study aimed to explore CD2AP's function in LUAD and its potential as a biomarker and therapeutic target through multi-omics analysis.
    METHODS: We analyzed CD2AP expression and clinical significance in LUAD using data from TCGA, GEO, and other public databases. We employed transcriptomics, methylation analysis, immune infiltration assays, and spatial transcriptomics. Kaplan-Meier survival analysis was used to assess the relationship between CD2AP expression and prognosis. Enrichment analysis identified biological processes and pathways related to CD2AP, while its association with the immune microenvironment and drug sensitivity was also evaluated.
    RESULTS: CD2AP was significantly overexpressed in LUAD, and high expression correlated with poorer prognosis, including overall survival and progression-free survival. Enrichment analysis showed CD2AP is involved in cell adhesion, PI3K-Akt signaling, and immune escape, suggesting it promotes LUAD progression through these pathways. High CD2AP expression was associated with alterations in the tumor immune microenvironment and drug sensitivity, particularly to chemotherapeutics like Cisplatin, Etoposide, and Paclitaxel, and resistance to targeted therapies like Gefitinib. Spatial transcriptomics revealed higher CD2AP expression in tumor regions, especially in malignant cell-enriched areas.
    CONCLUSION: This study highlights CD2AP's critical role in LUAD, particularly in immune microenvironment modulation, metabolic reprogramming, and drug response. CD2AP's high expression is linked to poor prognosis and may serve as a potential target for immunotherapy and drug response prediction.
    Keywords:   CD2AP ; Disulfidptosis; Drug sensitivity; LUAD; Multi-omics; Programmed cell death; Therapeutic target
    DOI:  https://doi.org/10.1007/s12672-025-02308-6
  7. Front Oncol. 2025 ;15 1543463
       Background: Lung adenocarcinoma (LUAD) is the primary subtype of Non-small cell lung cancer (NSCLC) and a serious threat to human health. However, the precise molecular mechanisms in lung cancer remain largely unexplored.
    Methods: Herein, we performed proteomic analysis in a cohort of 20 LC primary tumors and their paired normal tissues. The expression levels and prognostic value of hub proteins were also explored in LUAD using public databases. Glycinamide ribonucleotide transformylase (GART) expression was detected by qRT-PCR in LC cell lines. The roles of GART were assessed by CCK-8, colony formation, Wound healing assays, and xenograft tumor model. Expression levels of the PAICS-Akt-β-catenin pathway were estimated through qRT-PCR and western blot assays.
    Results: The proteomic analysis of tumor tissues of LC indicated that 263 proteins were upregulated and 194 were downregulated. Bioinformatics analysis showed that differentially expressed proteins were mainly associated with the regulation of apoptotic process and cell adhesion, PI3K-Akt signaling pathway, Purine metabolism, and Wnt signaling pathway. The expression of hub proteins EPRS, GART, HSPE1, and RPS6 was much higher in LUAD tissues than in normal tissues analyzed by the Ualcan database. Overexpression of GART represented a poor prognosis in LUAD patients. Additionally, the knockdown of GART effectively inhibited the cell proliferation and migration of LC cells both in vitro and in vivo. Mechanistically, qRT-PCR and western blot analyses suggested that GART deletion could inhibit the activation of the PAICS-Akt-β-catenin pathway in vivo.
    Conclusions: Our study indicated a tumor-promoting function of GART in LC through the regulation of the PAICS-Akt-β-catenin axis, and it may be used as a therapeutic target for NSCLC.
    Keywords:  GART; PAICS; cell migration; cell proliferation; lung cancer
    DOI:  https://doi.org/10.3389/fonc.2025.1543463
  8. Int J Surg. 2025 Apr 04.
       BACKGROUND: The relationship between the Advanced Lung Cancer Inflammation Index (ALI) and all-cause mortality in patients with Metabolic Dysfunction-Associated Steatohepatitis and Metabolic-Associated Alcoholic Liver Disease and other combination etiology of steatosis (MASLD/MetALD & Mixed Etiology Steatosis) is not well-understood. Current evidence is insufficient to establish this association, yet it holds critical importance for healthcare and public health. Research into the link between ALI and all-cause mortality in MASLD/MetALD & Mixed Etiology Steatosis remains a topic of interest.
    OBJECTIVE: This study investigated the association between ALI and all-cause mortality in MASLD/MetALD & Mixed Etiology Steatosis patients, and explored the clinical significance of this association.
    METHODS: We conducted a cohort study using data from the National Health and Nutrition Examination Survey between 2007 and 2018, involving 4502 adult participants with MASLD/MetALD & Mixed Etiology Steatosis in the United States. Data collected included age, sex, race, education, marital status, poverty-to-income ratio, alanine aminotransferase levels, aspartate aminotransferase levels, high-density lipoprotein cholesterol, total cholesterol, diabetes mellitus, coronary heart disease, and stroke. Cox proportional hazards regression models were used to assess the relationship between ALI and all-cause mortality, with follow-up through 31 December 2019, from the National Center for Health Statistics.
    RESULTS: The study found that ALI in patients was significantly negatively associated with the risk of all-cause mortality in U.S. adults with MASLD/MetALD & Mixed Etiology Steatosis. Participants with higher ALI levels had a significantly lower risk of all-cause mortality compared to those with lower ALI levels. After full adjustment, moderate ALI levels were associated with a 42% reduced risk (hazard ratio [HR]: 0.58, 95% confidence interval [CI]: 0.41-0.81), and high ALI levels were associated with a 49% reduced risk (HR: 0.51, 95% CI: 0.35-0.73) of all-cause mortality. No significant interactions were observed in subgroup analyses (P > 0.05).
    CONCLUSION: This study suggested that high ALI levels are associated with a reduced risk of all-cause mortality in MASLD/MetALD & Mixed Etiology Steatosis patients. These findings may have important clinical implications for healthcare providers managing MASLD/MetALD & Mixed Etiology Steatosis patients, emphasizing the potential role of ALI as a prognostic marker for all-cause mortality.
    Keywords:  metabolic dysfunction-associated steatohepatopathy (MASLD); metabolism-associated alcoholic liver disease (MetALD); prognostic assessment; the advanced lung cancer inflammation index (ALI)
    DOI:  https://doi.org/10.1097/JS9.0000000000002374
  9. Oncol Res. 2025 ;33(4): 863-872
      Lung cancer is one of the main causes of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) being the most prevalent histological subtype of lung cancer. Glutathione peroxidase 4 (GPX4) is a crucial antioxidant enzyme that plays a role in regulating ferroptosis. It is also involved in a wide variety of biological processes, such as tumor cell growth invasion, migration, and resistance to drugs. This study comprehensively examined the role of GPX4 in NSCLC and investigated the clinical feasibility of targeting GPX4 for NSCLC treatment. We discovered that GPX4 influences the progression of NSCLC by modulating multiple signaling pathways, and that blocking GPX4 can trigger ferroptosis and increase the sensitivity to chemotherapy. As a result, GPX4 represents a prospective therapeutic target for NSCLC. Targeting GPX4 inhibits the development of NSCLC cells and decreases their resistance to treatment.
    Keywords:  Biological function; Drug resistance; Glutathione peroxidase 4 (GPX4); Inhibitor; Non-small cell lung cancer (NSCLC)
    DOI:  https://doi.org/10.32604/or.2024.054201
  10. Cancer Cell Int. 2025 Apr 10. 25(1): 138
       BACKGROUND: Lung adenocarcinoma (LUAD) is the predominant form of non-small cell lung cancer (NSCLC). Mitochondrial quality-related genes (MQRGs) contribute to the genesis and advancement of tumors. Despite advances in LUAD treatment and detection, early diagnostic biomarkers are still lacking, and the roles of MQRGs in LUAD are not well understood.
    METHODS: We extensively examined transcriptome and clinical data from TCGA and GEO databases to discover differentially expressed MQRGs. Utilizing the LASSO algorithm and multivariate COX regression, a predictive risk model was created. Kaplan-Meier study and ROC curves were implemented to predict patient prognosis, resulting in a new Mitochondrial Quality Regulation Gene Signature for accurate prognosis forecasting. R software and packages facilitated statistical, consensus cluster, survival, Cox regression, Lasso regression, and tumor microenvironment analyses. Model-related gene expression was measured using RT-qPCR, immunohistochemistry, single-cell sequencing, HPA data, and UNCAN data.
    RESULTS: We created a concise risk model using four MQRGs (STRAP, SHCBP1, PKP2, and CRTAC1) to forecast overall survival in LUAD patients. High-risk patients experienced significantly lower survival rates. Functional analysis linked these MQRGs to alpha-linolenic acid metabolism pathways. Moreover, the tumor immune microenvironment supports previous findings that higher CD8 + T cell infiltration improves LUAD outcomes. Analysis of different risk scores showed increased activated memory T-cell CD4, suggesting its activation is crucial for LUAD prognosis. Nomograms were generated with clinical data and the MQRGscore model. mRNA and IHC analysis manifested significantly upregulated STRAP, SHCBP1, and PKP2 expression and mitigated CRTAC1 expression in the LUAD contrasted with normal lung tissue. qRT-PCR and immunohistochemistry confirmed these findings, aligning with TCGA data.
    CONCLUSIONS: We created a succinct MQRGs risk model to ascertain the LUAD patient's prognosis, potentially offering a novel method for diagnosing and treating this condition.
    Keywords:  LUAD; Mitochondrial quality regulation genes; Overall survival; Prognosis; Tumor microenvironment; Tumor mutation burden
    DOI:  https://doi.org/10.1186/s12935-025-03764-4
  11. Cancer Res. 2025 Apr 09.
      Chronic psychological stress is often associated with manifestations of malignant diseases. Identification of modulators regulating the interaction between stress and tumorigenesis could provide potential strategies to ameliorate cancer progression. Here, we observed that chronic stress markedly promoted lung cancer progression. Analysis of the landscape of lncRNA expression indicated that lncRNA HIF1A-AS3 was upregulated in the stressed group and in lung cancer specimens compared to normal tissues. HIF1A-AS3 promoted proliferation and invasion of lung cancer cells both in vitro and in vivo. Mechanistically, HIF1A-AS3 translationally activated HIF-1α via direct interaction with YBX1, stimulating downstream signaling cascades. HIF-1α inversely stimulated HIF1A-AS3 transcription by directly binding to its promoter region. Investigation of the immune microenvironment revealed that macrophage depletion could efficiently abolish the tumor promoting effects of chronic stress. Both chronic stress and HIF1A-AS3 overexpression induced M2-like macrophage polarization in tumor tissues in mice. Conditioned medium from HIF1A-AS3 overexpressing lung cancer cells enhanced the mobility and phagocytic activity of human and murine macrophages. Targeting HIF1A-AS3/HIF-1α signaling, which was aberrantly upregulated in human lung cancer specimens and predictive of poor prognosis, counteracted chronic stress-induced lung cancer progression in vivo. In conclusion, the HIF1A-AS3/HIF-1α positive feedback loop mediates chronic stress-induced lung cancer growth through functional reprogramming of tumor-associated macrophages, suggesting that this axis may serve as a promising diagnostic and therapeutic target for patients with lung cancer suffering from psychological stress.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-24-3481
  12. J Cancer Prev. 2025 Mar 30. 30(1): 47-55
      Chromosomal alterations are frequent events in lung cancer progression. Although gains and losses of chromosomal position have been reported, the association between copy number alteration and lung cancer patient survival has not been extensively investigated. In this study, we performed a meta-analysis of public cBioPortal datasets spanning 25 lung cancer studies to identify putative cancer driver genes with copy number alterations associated with overall patient survival. Ten copy-number altered genes enriched in deceased lung cancer patients were identified. Seven of these putative driver genes were located in the 7p11.2 chromosomal location, and two were in the 9p21.3 cytoband. Among these genes, the mitochondrial ribosomal protein S17 (MRPS17) amplification was significantly associated with a lower patient survival rate (P = 1.47e-7). To investigate the functional role of MRPS17, small interfering RNA-mediated knockdown was performed in two non-small cell lung cancer cell lines, A549 and NCI-H460. MRPS17 knockdown significantly reduced cell proliferation, migration, invasion, and anchorage-independent growth in both cell lines. Furthermore, knockdown of MRPS17 decreased the activation of the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, suggesting its role in driving lung cancer progression through this critical oncogenic pathway. Our findings highlight MRPS17 as a potential cancer therapy target and a prognostic biomarker that may improve the survival rates of lung cancer patients. Future studies should explore its inhibition as a therapeutic strategy as well as elucidate its molecular mechanisms in cancer progression.
    Keywords:  Biomarker; Lung cancer; Mitochondrial ribosomal protein S17; Patient survival; Small interfering RNA
    DOI:  https://doi.org/10.15430/JCP.24.023
  13. Redox Biol. 2025 Mar 23. pii: S2213-2317(25)00123-5. [Epub ahead of print]82 103610
      The efficacy of strategies targeting oncogenic RAS, prevalent in lung adenocarcinoma (LUAD), is limited by rapid adaptive resistance mechanisms. These include loss of RAS addiction and hyperactivation of downstream signaling pathways, such as PI3K/AKT. We previously reported that oncogenic RAS-driven LUAD cells possess an enhanced reliance on MTH1, the mammalian 8-oxodGTPase, to prevent genomic incorporation of oxidized nucleotides, and that MTH1 depletion compromises tumorigenesis and oncogenic signaling. Here, we show that elevated MTH1 correlates with poor prognosis in LUAD and that its redox-protective 8-oxodGTPase activity is variably regulated in KRAS-addicted vs. non-addicted states. Multiple oncogenic KRAS mutants or overexpression of wildtype (wt) KRAS increased MTH1 expression. Conversely, KRAS depletion or its inhibition by AMG-510 (sotorasib) decreased MTH1 in KRASG12C-addicted LUAD cells. Separation-of-function MEK/ERK1/2-activating mutants recapitulated the elevated MTH1 expression induced by oncogenic RAS in wt KRAS LUAD cells. However, upon inhibition of the MEK/ERK1/2 pathway, compensatory AKT activation maintained MTH1 expression. Indeed, elevated AKT signaling maintained high MTH1 expression even when KRAS oncoprotein was low. We previously reported that cancer cells possess variable MTH1-specific and MTH1-independent 8-oxodGTPase activity levels. Whereas both ERK1/2 and AKT could regulate MTH1 protein levels in KRAS-addicted cells, only AKT signaling was associated with elevated MTH1-specific 8-oxodGTPase activity under KRAS-low or KRAS non-addicted states. Our studies suggest that despite loss of KRAS dependency, LUAD cells retain the requirement for high MTH1 8-oxodGTPase activity due to redox vulnerabilities associated with AKT signaling. Thus, MTH1 may serve as a novel orthogonal vulnerability in LUAD that has lost KRAS addiction.
    Keywords:  8-Oxo-dGTPase; AKT signaling; ERK signaling; KRAS inhibition; MTH1; NUDT1; Oncogene addiction; Redox-protective adaptations
    DOI:  https://doi.org/10.1016/j.redox.2025.103610
  14. Medicine (Baltimore). 2025 Apr 04. 104(14): e42053
      Lung cancer ranks highest in annual mortality among all cancers, and blood metabolites may influence its onset and progression. Our objective is to assess the causal relationships between blood metabolites and both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), while exploring the mediating effects of immune cells. We utilized publicly available genetic data to investigate the potential causal relationships between blood metabolites and NSCLC as well as SCLC using a 2-sample Mendelian randomization (MR) approach. In our primary analysis, we employed the inverse variance weighted (IVW) method and conducted sensitivity analyses and Steiger test to assess the reliability and directionality of the causal relationships. Additionally, we employed a 2-step MR approach to evaluate the mediating role of immune cells in these causal relationships. The IVW method revealed that palmitoylcarnitine levels (metabolon platform) and 4 other blood metabolites are risk factors for NSCLC, while tetrahydrocortisol glucuronide levels and 2 other blood metabolites are protective factors for NSCLC. Additionally, Alpha-hydroxyisovalerate levels and 8 other blood metabolites are risk factors for SCLC, whereas dimethylglycine levels and 3 other blood metabolites are protective factors for SCLC. Furthermore, IgD- CD27- B cell %B cell, CD27 on IgD- CD38dim B cell, and CD3 on Naive CD4+ T cell mediate some of the relationships between blood metabolites and NSCLC. Activated and secreting CD4 regulatory T cell %CD4+ T cell, CD14- CD16- Absolute Count, and IgD on IgD+ CD24+ B cell mediate some of the relationships between blood metabolites and SCLC. There are significant causal relationships between blood metabolites and both NSCLC and SCLC, with some of these relationships mediated by immune cells. This aids us in influencing the role of blood metabolites in lung cancer by intervening with immune cells, thereby providing more avenues for the prevention and treatment of lung cancer.
    Keywords:  Mendelian randomization; blood metabolites; immune cells; non-small cell lung cancer; small cell lung cancer
    DOI:  https://doi.org/10.1097/MD.0000000000042053