ANXA1 inhibits ferroptosis in HUVECs via negative regulation of ferritinophagy
Ferroptosis is a phospholipid peroxidation-mediated and iron-dependent cell death form. Inhibiting ferroptosis is a promising strategy for the prevention and treatment of cardiovascular diseases. Annexin A1 (ANXA1) is an endogenous anti-inflammatory mediator that plays an important regulatory role in cardiovascular diseases such as atherosclerosis, especially in inflammation suppression, protecting the heart from injury, and regulating vascular function. Ac2-26 is a synthetic peptide derived from the N-terminal 26 amino acids of ANXA1, which retains its anti-inflammatory properties. However, the regulatory mechanism of ANXA1 in atherosclerosis (AS) is not yet fully understood. This study aims to explore the specific role of ferroptosis in HUVECs and demonstrate that ANXA1 can disrupt ferritinophagy and protect endothelial function. Treatment with Ac2-26 or ANXA1-overexpressing HUVECs alleviated RSL3-induced endothelial cell dysfunction and inhibited lipid peroxidation, as evidenced by a reduction in ferrous ion levels and upregulation of GPX4, FTH1, and SLC7A11 protein expression, along with a downregulation of LC3-II and NCOA4 expression. In contrast, knockdown of ANXA1 in HUVECs failed to suppress the outcome of ferroptosis. Furthermore, co-immunoprecipitation analysis revealed that ANXA1 overexpression prevented ferritin degradation by disrupting the NCOA4-FTH1 protein-protein interaction, reducing the bioavailability of intracellular ferrous ions, thereby blocking ferroptosis. In conclusion, our findings identify a novel mechanism, showing that ANXA1 can inhibit ferroptosis via ferritinophagy, thereby alleviating endothelial dysfunction, which may provide a new therapeutic avenue for AS.
miR-98-3p/VEGFA axis mediates MALAT1-induced angiogenesis in ovarian tumors
The functional role of MicroRNA miR-98-3p in ovarian cancer is largely unexplored and its molecular mechanisms remain incompletely understood. In this study, we identified a novel regulatory axis involving MALAT1, miR-98-3p, and VEGFA in ovarian cancer angiogenesis. The study focuses on ovarian cancer-related proliferation and migration effects, primarily involving the angiogenesis effects of ovarian cancer. RNA sequencing following MALAT1 knockdown in HEY-T30 cells revealed significant alterations in several miRNAs, particularly miR-98-3p. Luciferase reporter assays confirmed direct binding between MALAT1 and miR-98-3p, establishing MALAT1 as a competing endogenous RNA (ceRNA) for miR-98-3p. Bioinformatic analysis and luciferase reporter assays further identified VEGFA as a direct target of miR-98-3p. Clinical database analysis demonstrated a positive correlation between MALAT1 and VEGFA expression, with elevated levels of both being significantly associated with poor overall survival in ovarian cancer patients. Functionally, both MALAT1 knockdown and miR-98-3p overexpression significantly impaired HUVEC tube formation, proliferation, and migration, which could be reversed by miR-98-3p inhibition. In vivo, miR-98-3p overexpression in subcutaneous xenografts resulted in reduced tumor volume, weight, vasculature, and blood perfusion, along with decreased expression of VEGFA, MMP2, and MMP9. These findings elucidate a MALAT1/miR-98-3p/VEGFA regulatory axis that modulates tumor angiogenesis in ovarian cancer, providing potential therapeutic targets for this malignancy.
SLBP Promotes Lung Adenocarcinoma Progression by Inhibiting Ferroptosis and Reprogramming Glutamine Metabolism via FADS2 Interaction
SLBP is significantly overexpressed in lung adenocarcinoma (LUAD) and correlates strongly with poor patient prognosis. Functional studies demonstrated that SLBP potently enhances proliferation, invasion, and metastasis of LUAD cells both in vitro and in vivo. Mechanistically, SLBP suppresses ferroptosis-a form of regulated cell death-by modulating key biochemical markers, including GSH, MDA, and Fe levels, and it restores cell viability upon treatment with ferroptosis inducers. RNA-seq and biochemical analyses revealed that SLBP transcriptionally upregulates and stabilizes SLC7A11, a critical ferroptosis suppressor, thereby inhibiting lipid peroxidation and ferroptotic cell death. Additionally, immunopurification and mass spectrometry (IP-MS) identified FADS2 as a novel SLBP-interacting partner. SLBP binds to FADS2, promotes its expression, and drives metabolic reprogramming toward glutamine dependency, significantly altering choline and metal ion metabolism. This metabolic shift enhances cellular proliferation under nutrient stress. Crucially, SLBP-mediated proliferation was shown to be functionally dependent on FADS2, as FADS2 inhibition abrogates SLBP-driven growth without affecting SLBP levels. Collectively, these results uncover SLBP as a multifunctional oncoprotein that promotes LUAD progression through dual mechanisms: inhibiting ferroptosis via SLC7A11 and rewiring glutamine metabolism through FADS2, offering new potential targets for therapeutic intervention.
IL-33/ST2 signaling promotes intrahepatic cholangiocarcinoma through reprogramming macrophage polarization via MAPK pathway
The role of IL-33/ST2 signaling in reprogramming macrophage polarization within the intrahepatic cholangiocarcinoma (ICC) microenvironment remains poorly understood. This study aimed to elucidate the mechanisms by which IL-33/ST2 signaling regulates macrophage polarization and its impact on ICC progression.
Effects of everolimus on circadian gene expression and cell fate in synchronized Caco-2 cells
Circadian rhythms regulate key biological processes, including cell proliferation and metabolism, and their disruption is implicated in colorectal cancer (CRC). mTOR signaling interacts bidirectionally with the circadian clock, yet how mTOR inhibition modulates clock gene dynamics and cellular behavior in intestinal models remains unclear. This study aimed to investigate the effects of everolimus, an mTOR inhibitor, on circadian gene expression, cell viability, apoptosis, and cell cycle progression in synchronized Caco-2 cells, with consideration of cell confluency and Circadian Time (CT).
MicroRNA-124-3p suppresses lung cancer by targeting ITGB1/PI3K/p-AKT signal transduction pathway
Worldwide, lung cancer is a leading cause of cancer-related death, and non-small cell lung cancer (NSCLC) represents the most common histological subtype. Numerous studies have demonstrated that microRNAs (miRNAs) play critical roles in NSCLC pathogenesis. microRNA-124-3p (miR-124-3p) has been identified as a tumor-associated miRNA, and we confirmed its downregulation in NSCLC cells. Functional assays showed that overexpression of miR-124-3p suppresses proliferation and migration of NSCLC cells, whereas its knockdown promotes these malignant phenotypes. A luciferase reporter assay revealed that miR-124-3p directly targets ITGB1 by binding to its 3'-UTR. Mechanistically, ITGB1 enhances PI3K expression and increases AKT phosphorylation, thereby activating the PI3K/AKT signaling pathway. Notably, miR-124-3p retained its tumor-suppressive effects even in A549 cells engineered to express the PIK3CA mutation. Consistent with this, bioinformatics analysis demonstrated that miR-124-3p expression is significantly lower in tumor tissues than in adjacent normal lung and further decreases in advanced T stage (T3-T4) compared to early stage (T1-T2). These findings indicate that miR-124-3p inhibits NSCLC progression via the ITGB1/PI3K/p-AKT axis and remains functional despite PIK3CA activation, supporting its potential as a therapeutic candidate.
MicroRNA-Mediated Metabolic Control in HCC: From Molecular Networks to Therapy
Hepatocellular carcinoma (HCC) is the most common histological subtype of primary liver cancer, accounting for nearly 80-90% of all liver cancer cases. While 'liver cancer' refers broadly to all malignant tumors arising in the liver (including HCC, cholangiocarcinoma, and others), HCC specifically originates from hepatocytes. HCC is characterized by extensive metabolic reprogramming, including not only higher levels of aerobic glycolysis, de novo lipogenesis, and altered glutamine metabolism but also altered one-carbon metabolism. Enhanced metabolic adaptation markers, therefore, serve as key indicators of malignant transformation but also contribute to cancer progression by promoting cell proliferation, metastasis, immune modulation, and therapy evasion. Emerging evidence suggests microRNAs (miRNAs) can coordinate these metabolic adaptations by targeting key enzymes, transporters, transcription factors, signaling molecules, or pathways involved in metabolism. For instance, miR-122, miR-27a, miR-148a, and miR-4310 inhibit lipid accumulation and mitochondrial dysfunction, while miR-21, miR-103a, and miR-30b-5p promote glycolysis, lipogenesis, and anabolic metabolism. Long non-coding RNAs (lncRNAs) and exosomal miRNAs interact with these upstream regulators to form a heterogeneous network of non-coding RNAs. These networks participate in remodeling the tumor microenvironment, modulating the immune response, and facilitating metabolic adaptation in HCC. miRNAs are ideal for the potential stratification of HCC risk, prognosis, and therapeutic response, as they occupy key upstream positions in regulatory hierarchies and have been described as both biomarkers and potential metabolic switches. This study aims to elucidate the role of microRNAs in regulating metabolic pathways in HCC by delineating numerous miRNA-target interactions involved in glycolysis, lipid, amino acid, and nucleotide metabolism. This knowledge will enable us to identify novel diagnostic biomarkers and therapeutic targets for prognosis and to explore effective novel precision treatment strategies.
Sagittaria sagittifolia polysaccharide regulates Nrf2-mediated antioxidant to improve apoptosis and ferroptosis in high glucose-induced lens epithelial cells
Diabetic cataract (DC), a primary ocular complication of diabetes mellitus, remains a leading cause of global blindness. The bioactive polysaccharide Sagittaria sagittifolia polysaccharide (SSP) exhibits remarkable antioxidant and anti-apoptotic efficacy in age-related cataract models, yet its DC therapeutic potential is unexplored. This study investigated SSP's protective effects against high glucose (HG)-induced damage across three experimental models, focusing on nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant in apoptosis and ferroptosis regulation. In ex vivo rat lenses, SSP pretreatment (1 mg/mL, 24 h) significantly attenuated HG-induced (25 mM, 48 h) lens opacification. In human lens epithelial B3 (HLEB3) cells, SSP pretreatment (1 mg/mL, 24 h) markedly improved cell viability and proliferation under HG conditions (150 mM, 72 h). Mechanistically, SSP significantly decreased oxidative stress markers (malondialdehyde, protein carbonyls, reactive oxygen species) while restoring mitochondrial function and enhancing antioxidant capacity (glutathione levels, catalase activity). SSP activated the Nrf2 pathway, regulating key antioxidant proteins (NAD(P)H quinone dehydrogenase 1, heme oxygenase-1, thioredoxin (Trx), Trx2 and glutaredoxin 1 to mitigate HG-induced oxidative damage. SSP exerted anti-apoptotic effects by upregulating B-cell lymphoma-2 (Bcl-2) while suppressing Bcl-2-associated X and cleaved caspase-3 expression. SSP modulated ferroptosis by increasing Ferritin, system Xc, and glutathione peroxidase 4 (GPX4) while reducing Fe and acyl-CoA synthetase long-chain family member 4 (ACSL4) levels. In STZ-induced diabetic mice, SSP treatment ameliorated lens epithelial cells (LECs) morphological damage and reduced protein expression of caspase-3 and ACSL4, whereas increased protein expression of GPX4 and ferritin. Crucially, upon Nrf2 knockdown in HLEB3 cells via short interfering RNA, SSP confirmed its protective role by activating Nrf2 to inhibite apoptosis and ferroptosis. Collectively, these findings demonstrate that SSP protects LECs against HG-induced damage through Nrf2-mediated coordination of antioxidant defense, anti-apoptotic, and anti-ferroptotic mechanisms, highlighting its therapeutic potential for DC.
lincRNA-p21 drives apoptosis and calcification of vascular smooth muscle cell via small extracellular vesicles under hyperphosphatemic conditions in chronic kidney disease
Vascular calcification, a major contributor to cardiovascular morbidity, involves pathological osteogenic transdifferentiation of vascular smooth muscle cell (VSMC) under hyperphosphatemic conditions in chronic kidney disease. This study investigates the role of lincRNA-p21 and small extracellular vesicles in phosphate-induced mouse aortic smooth muscle cell (MASMC) calcification. Exposure to phosphate (2.6 mmol/L) triggered time-dependent calcification, characterized by enhanced calcium deposition, endoplasmic reticulum remodeling, and intracellular calcium accumulation (p < 0.05 vs. 24/48-h controls). Concurrently, the expression of osteogenic markers (BGP, OCN, and OPN) and lincRNA-p21 was significantly upregulated, whereas the expression of contractile phenotype-specific markers (SM22α, SM-MHC, and SM α-actin) was markedly down-regulated. This pattern of gene expression was correlated with MASMC osteogenic transdifferentiation. Small extracellular vesicles isolated from phosphate-treated MASMC exhibited elevated lincRNA-p21 levels (p < 0.05) and induced calcification and apoptosis in recipient cells, suggesting small extracellular vesicles-mediated propagation of calcific signals. Functional studies demonstrated that lincRNA-p21 overexpression exacerbated calcification, apoptosis, and osteogenic marker expression, while its knockdown attenuated these effects (p < 0.05). Time-course analyses revealed lincRNA-p21 dynamically regulates small extracellular vesicles secretion, calcium accumulation, and apoptotic pathways, acting as a molecular switch driving phosphate-induced calcification. These findings establish lincRNA-p21 as a critical mediator of MASMC calcification via small extracellular vesicles-dependent mechanisms, offering insights into therapeutic strategies for vascular calcification.
Mulberry leaf flavonoid extract inhibits high glucose-induced ferroptosis by regulating the PI-3K/Akt/Nrf2/GPX4 pathway in HT22 mouse hippocampal neuronal cells
To explore the inhibitory effects of mulberry leaf flavonoid extract (MFE) on high glucose-induced diabetic encephalopathy (DE) in a cell model, and to investigate the underlying mechanisms combined with network pharmacology prediction and experimental validation.
Augmenter of liver regeneration alleviates lipid accumulation in MASLD through ATF3-CD36 axis
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by excessive lipid deposition in hepatocytes. Augmenter of liver regeneration (ALR, encoded by the Gfer gene), a survival factor in liver cells, protects the liver against various injuries, whereas downregulation of ALR expression accelerates MASLD progression in mice. CD36 functions as a long chain free fatty acid transporter, playing a crucial role during pathogenesis of MASLD. Herein, the expression level of CD36 was found to significantly decrease in ALR-transfected (ALR-Tx) HepG2 cells and upregulated in shRNA-ALR (shALR) cells after oleic acid/palmitic acid treatment. Furthermore, CD36 expression was markedly elevated in MASLD model mice with heterozygous Gfer knockdown (Gfer) fed a high-fat diet,as well as model mice with liver-specific Gfer knockout (Gfer-CKO). Lipid uptake increased and lipophagy was suppressed in shALR HepG2 cells, and conversly, lipid uptake reduced and lipophagy was promoted in ALR-Tx HepG2 cells. After downregulation of CD36 expression by siRNA-CD36 in shALR cells, lipid accumulation markedly decreased, and lipophagy was stimulated. Further, the mRNA and protein expression of ATF3 (Activating Transcription Factor 3) were found to substantially elevated and decreased in ALR-Tx cells and shALR cells respectively, and CD36 expression was induced after ATF3 was knock down. Our results thus suggest that lack of ALR aggravates lipid deposition in MASLD partly through the ATF3-CD36 axis.
Timeless prevents senescence-associated phenotypes and enhances DNA repair to promote esophageal cancer cell growth
Esophageal squamous cell carcinoma (ESCC) is one of the deadliest forms of squamous cell carcinoma, comprising approximately 90% of all esophageal cancer cases. We previously demonstrated that the Fanconi anemia DNA repair (FA) pathway mitigates replication stress to preserve the self-renewal capacity of esophageal cancer cells, highlighting the critical role of minimizing replication stress in esophageal cancer proliferation. In this study, to further explore the role of replication stress in esophageal cancer growth, we investigated the function of Timeless, a key subunit of the replication fork protection complex essential for preventing replication stress. Our findings reveal that Timeless is upregulated in esophageal cancer cells, and its depletion increases sensitivity to DNA-damaging agents, inducing cellular senescence in esophageal keratinocytes. Timeless depletion also elevates the DNA damage response while reducing the expression of DNA repair proteins associated with the FA pathway and homologous recombination. Furthermore, the loss of Timeless impairs colony-forming ability in soft agar and diminishes the self-renewal capacity required to form 3D organoids. These results suggest that Timeless plays a critical role in facilitating DNA repair and esophageal cancer progression and may represent a promising target for developing effective therapeutic strategies to treat esophageal cancers.
Metformin inhibits heterotopic ossification of mouse Achilles tendon by inhibiting the Nr4a1/Wnt/β-catenin signaling pathway
Heterotopic ossification is a pathological process characterized by aberrant bone formation in soft tissues, leading to joint pain, stiffness, functional impairment, and poor quality of life. We used a mouse Achilles tendon heterotopic ossification model and in vitro tendon-derived stem cells (TDSCs) assays to identify the molecular mechanism of metformin in prevention of heterotopic ossification. Metformin significantly attenuated heterotopic ossification, reducing ectopic bone volume and osteogenic gene expression. In vitro, metformin inhibited TDSCs osteogenic differentiation in a dose-dependent manner, decreasing calcium nodule deposition and osteogenic marker expression. Transcriptomic analysis revealed downregulated Nr4a1 expression in metformin-treated heterotopic ossification samples; in vitro experiments confirmed that Nr4a1 activation enhances TDSCs osteogenesis, and Nr4a1 knockdown suppresses osteogenesis. Metformin also reduced Wnt4 and β-catenin expression, suggesting that Nr4a1 promotes heterotopic ossification by positively regulating Wnt/β-catenin signaling. In sum, metformin downregulates expression of Nr4a1 in TDSCs, which suppresses osteogenic differentiation by inhibiting Wnt/β-catenin signaling. As a mediator of TDSC osteogenic differentiation, Nr4a1 may be a therapeutic target for heterotopic ossification.
Elucidating the impact of trans-ned-19 on two-Pore channel 2 mutants of Dictyostelium: changes in intracellular calcium levels and subsequent effect on autophagic flux
Two-Pore Channel 2 (TPC2) are calcium (Ca) release channels regulated by Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP), primarily localized to the endolysosomal system. TPC2 regulates diverse Ca-dependent processes such as cell proliferation, development, migration, and autophagy. In the protist Dictyostelium discoideum, intracellular Ca levels and autophagy are key determinants of cell-fate, particularly for stalk cell differentiation. To investigate the involvement of DdTPC2 in NAADP-mediated Ca signalling, we treated wild-type (Ax2) and various tpc2 mutant strains, like overexpressors (tpc2), knockout (tpc2), and rescue (tpc2), with both NAADP-AM (NAADP agonist) and trans-Ned-19 (NAADP antagonist). Our findings show that trans-Ned-19 does not specifically inhibit DdTPC2 activity during the proliferative (vegetative) stage, but acts as a specific TPC2 inhibitor in freshly starved cells or during multicellular development, as the phenotypes displayed by trans-Ned-19 treated Ax2 cells were similar to tpc2 cells. Trans-Ned-19 colocalizes with lysosomes, consistent with the subcellular distribution of TPC2. Fluid-phase endocytosis was reduced in tpc2 cells, implicating TPC2 in endolysosomal function. Intracellular calcium measurements demonstrated that NAADP regulates Ca signalling through TPC2, as both NAADP-AM and trans-Ned-19 significantly modulated intracellular free Ca levels in TPC2-expressing strains. Notably, trans-Ned-19 treatment reduced autophagic flux in Ax2 cells to tpc2 levels, demonstrating a positive correlation between TPC2 and autophagic flux, further supported by rapamycin and 3-methyladenine treatment results. This study highlights TPC2 as a pivotal regulator of NAADP-mediated Ca signalling and autophagy in Dictyostelium, with broad implications for understanding these processes in higher eukaryotes.
Fbxo45 promotes cell viability, invasion and sunitinib resistance of clear cell renal cell carcinoma by targeting Erbin
The mechanisms underlying the development and progression of clear cell renal cell carcinoma (ccRCC) and its sunitinib resistance are elusive. Fbxo45 is a member of the F-box protein family that has been demonstrated to participate in tumorigenesis. However, the role of Fbxo45 in ccRCC progression has not been characterized. This study aims to investigate the biological functions and molecular mechanism of Fbxo45 in ccRCC progression. We found that Fbxo45 knockdown inhibited the viability and motility of ccRCC cells, while Fbxo45 overexpression resulted in the opposite phenotype. Ectopic expression of Fbxo45 promoted tumor growth in mice. Fbxo45 expression was negatively correlated with Erbin expression, which has been reported to mediate anti-tumor activities in ccRCC. Furthermore, Fbxo45 facilitated ccRCC cell viability and motility by inhibiting Erbin. Notably, Fbxo45 upregulation reduced sunitinib sensitivity in ccRCC cells. Our results suggest that Fbxo45 could be a potential target for ccRCC treatment and sunitinib resistance.
Glycolysis inhibition and AMPK activation: a critical role of CTRP1 deficiency in the treatment of hypoxia-induced pulmonary hypertension
In pulmonary hypertension (PH), metabolic enhancement of glycolysis drives a hyperproliferative and apoptosis-resistant phenotype in pulmonary artery smooth muscle cells (PASMCs), which is a key pathological process leading to pulmonary vascular remodeling. We aimed to uncover the role of CTRP1, a gene responsible for regulating glycolysis, in the modulation of PH-related pathogenesis. In the PH mouse and cell models established by hypoxia stimulation, the regulatory mechanism of CTRP1 deletion on the pathological characteristics of PH was explored. CTRP1 levels were significantly upregulated in PH mice, accompanied by an abnormal increase in lactate production and glycolysis-related key protein expressions (HK2 and PDHK1). Inhibition of CTRP1 markedly improved pulmonary artery pressure and right ventricular function in PH mice by reducing glycolysis levels. In vitro experiments further observed that CTRP1 knockdown suppressed the hypoxia-induced hyperproliferation and anti-apoptosis phenotype of PASMCs, with inhibition of glycolysis. Mechanically, downregulated CTRP1 resulted in p-AMPK activation and p-AKT/mTOR inhibition. This beneficial effect was reversed by AMPKα2 deficiency. Overall, CTRP1 deficiency reverses the hypoxia-induced hyperproliferation and antiapoptotic capacity of PASMCs by weakening glycolysis. These results provide evidence for CTRP1 as a potential therapeutic target in PH.
Pseudane V alleviates ox-LDL-induced macrophage M1 polarization by inhibiting mA modification of PPARGC1A
Atherosclerosis is driven by oxidized low-density lipoprotein (ox-LDL)-triggered macrophage malfunction, yet the precise pathways and effective counter-measures remain elusive. Here we delineated how N6-methyladenosine (mA) RNA methylation governs ox-LDL-induced M1 macrophage polarization and evaluated marine natural products for therapeutic intervention. Human monocytic THP-1 cells differentiated into M0 macrophages were treated with ox-LDL. M1/M2 polarization states were analyzed using flow cytometry, and changes in polarization markers were examined using quantitative real-time polymerase chain reaction (qRT-PCR). Global m6A changes were detected using mA dot blot and quantification analysis. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were performed to identify downstream target genes of the ox-LDL-mA pathway. The role of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) in ox-LDL-induced M1 polarization was analyzed. A high-throughput marine natural product library was employed to discover agents capable of reversing ox-LDL-driven macrophage polarization. ox-LDL induces macrophage M1 polarization, and M1 macrophages damage endothelial cells. ox-LDL-induced polarization of macrophages into the M1 phenotype was associated with increased global RNA mA modification. High levels of mA modification induced by ox-LDL suppressed PPARGC1A expression in a YTH domain family 2 (YTHDF2)-dependent manner, leading to M1 polarization. Pseudane V, a marine natural product, effectively reduced ox-LDL-induced M1 polarization and protected vascular endothelial cells by correcting abnormal mA modification of PPARGC1A. In conclusion, ox-LDL induces mA modification of PPARGC1A, suppressing its expression and promoting M1 polarization of macrophages, contributing to atherosclerosis development. Pseudane V counteracts excessive mA modification caused by ox-LDL, preventing M1 polarization of macrophages. These findings suggest the potential use of Pseudane V in preventing atherosclerosis.
The Aurora B inhibitor ZM-447439 induces caspase-independent necrosis-like death in v-Src oncogene-expressing cells via accumulation of extra-lysosomal cathepsin B
Src signaling is aberrantly activated in various cancers, and therapeutic strategies targeting Src-dependent cancers have been developed. We previously reported that cells expressing the oncogenic mutant v-Src are resistant to microtubule-targeting agents (MTAs), yet paradoxically exhibiting sensitivity to the Aurora B inhibitor ZM-447439; however, the mechanism underlying this cytotoxicity remains unclear. In this study, we demonstrate that the Aurora B inhibitor ZM-447439 potentiates cell death in v-Src-expressing HeLa S3 and HCT116 cells, accompanied by the accumulation of tetraploid and polyploid cells. The pan-caspase inhibitor z-VAD-FMK treatment does not affect the cell death. Time-lapse imaging analysis reveals cell death with the feature of necrosis in ZM-447439-treated v-Src expressing cells: altered morphology and loss of membrane integrity without chromatin condensation. Although autophagic flux is impaired, the autophagosome accumulation does not contribute to cell death. Notably, the cathepsin B expression is elevated in v-Src-expressing cells, and the amount of mature active cathepsin B outside lysosomes is strongly increased upon ZM-447439 treatment in v-Src-expressing cells. Treatment with the cathepsin B inhibitor CA-074 methyl ester mitigates cell death, similar to the pan-cysteine cathepsin inhibitor E64d treatment. These results suggest that the Aurora B inhibitor ZM-447439 potentiates caspase-independent necrosis-like death in v-Src-expressing cells partly through the accumulation of extra-lysosomal cathepsin B. The Aurora B inhibitors might be promising therapeutic agents for Src-driven cancers.
Histone H3 lysine 18 lactylation promotes alveolar epithelial cell apoptosis in sepsis-induced lung injury by upregulating caspase-8 in vivo and in vitro
Sepsis is a leading cause of acute lung injury (ALI), with apoptosis of alveolar epithelial cells (AECs) playing a central role. Elevated lactate, a hallmark of sepsis-induced metabolic reprogramming, has recently been implicated in lysine lactylation. However, its contribution to AEC apoptosis and ALI pathogenesis remains unclear.
Ferroptosis modulates invasion and migration in prostate cancer PC-3M subclones
Prostate cancer (PCa) metastasis remains a formidable clinical challenge, underscoring the urgent need to uncover its underlying molecular mechanisms. Leveraging the PC-3M cell line and its sublines with divergent metastatic potentials (PC-3M-1E8, highly metastatic; PC-3M-2B4, low-metastatic), this study investigated the role of ferroptosis, a form of iron-dependent regulated cell death, in PCa metastasis. Functional assays demonstrated that PC-3M-1E8 cells exhibited significantly enhanced proliferation, migration, invasion, and clonogenic capacity compared to PC-3M-2B4 cells. Proteomic analysis identified 5502 differentially expressed proteins, with GSEA pinpointing ferroptosis as the most significantly enriched pathway (NES = 2.8, FDR < 0.001) in PC-3M-1E8 cells. Key ferroptosis regulators, including GPX4 (1.4-fold), GCLM (1.6-fold), GCLC (1.5-fold), and TFRC (1.8-fold), were upregulated in PC-3M-1E8, while ACSL4 (0.7-fold) and VDAC3 (0.6-fold) were downregulated, constructing a ferroptosis-resistant network. Functional validation revealed that PC-3M-1E8 cells were more resistant to ferroptosis induction, evidenced by 3.68-fold higher GPX4 expression, 1.37-fold elevated GSH/GSSG ratios, and blunted iron responses. These findings establish an inverse correlation between ferroptosis sensitivity and PCa metastatic potential, providing a novel perspective on PCa metastasis by linking iron metabolism reprogramming to metastatic competence. Clinically, the identified ferroptosis-related signatures offer potential as predictive biomarkers for metastatic risk, and ferroptosis induction emerges as a promising therapeutic strategy for metastatic PCa. Future research should focus on exploring the crosstalk between ferroptosis and other cancer-related pathways to develop more effective targeted therapies.
microRNA-100 inhibits cell division in human non-small cell lung cancer via a CDC25A signaling mechanism
Lung cancer is one of the most common malignancies worldwide, and the prognosis remains unsatisfactory with conventional treatments. Our research revealed that miR-100 is significantly decreased in non-small cell lung cancer (NSCLC) tissues and cell lines, indicating its potential role in NSCLC development. The upregulation of miR-100 expression inhibited the growth of L18 NSCLC cells, while its downregulation increased the growth of A549 NSCLC cells. Cell cycle dysregulation is a key factor in NSCLC development. CDC25A is a vital cell cycle regulator that promotes cell cycle progression. Our objective was to determine the function of the CDC25A protein and its effect on the prognosis of NSCLC patients. The luciferase reporter gene assay demonstrated direct targeting of the 3' non-coding sequence of CDC25A by miR-100-3p. Western blot analysis confirmed that the protein expression levels of cyclin D1, CDK6, pRb, and E2F3 decreased upon miR-100 overexpression. Furthermore, cell cycle analysis demonstrated that miR-100 overexpression led to G1 phase arrest and a concomitant reduction in S phase entry in NSCLC cells. In vivo studies using a xenograft mouse model demonstrated that miR-100 overexpression inhibited tumor growth, while its inhibition accelerated it. Rescue experiments confirmed that CDC25A partially reversed the cell cycle arrest and growth suppression mediated by miR-100. This study's findings suggest that miR-100 can inhibit NSCLC progression by specifically targeting CDC25A, a cell cycle regulator, and its downstream molecular targets. Hence, miR-100 may have significant therapeutic potential against NSCLC.
