Published in last 50 years
Articles published on Human Cancer
- New
- Research Article
- 10.1055/a-2715-0775
- Nov 7, 2025
- Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme
- María M Adeva-Andany + 5 more
An independent association between insulin resistance and cancer has been consistently reported in humans. Patients with cancer display insulin resistance or its clinical manifestations, and this metabolic adaptation precedes the clinical diagnosis of cancer. Insulin resistance in cancer patients is associated with a metabolic switch from oxidative metabolism toward glycolysis that spares oxygen to be used in anabolic processes and facilitates the fast production of energy and intermediate metabolites required for the rapid proliferation of cancer cells. In malignant cells, glucose consumption via glycolysis occurs under normoxic conditions (aerobic glycolysis). Pathogenic mechanisms underlying insulin resistance in cancer patients include hypoxia-inducible factor-1 upregulation and overproduction of cytokines, such as interferon, interleukin-6, interleukin-18, and interleukin-1β. Deficit of 2-oxoglutarate (α-ketoglutarate) has been detected in cancer cells and may facilitate hypoxia-inducible factor-1 assembly and activity. Overproduction of cytokines in cancer patients follows activation of the immune system by abnormal nucleic acid variants. Anomalous DNA or RNA structures are recognized by immune sensors and stimulate signaling pathways that ultimately increase cytokine production. Likewise, interferon overproduction occurs in congenital disorders that feature ineffectively repaired DNA lesions, such as Werner syndrome, Bloom syndrome, mutations in DNA polymerase-δ1, and ataxia telangiectasia. These diseases cause simultaneous insulin resistance and a high tendency to develop cancer, highlighting the relationship between the two processes. Defectively repaired DNA injury endangers genomic integrity, predisposing to cancer, and activates the immune system to increase interferon production and subsequent insulin resistance. Hypoxia-inducible factor-1 and cytokines induce insulin resistance by suppressing peroxisome proliferator-activated-γ in the subcutaneous adipose tissue.
- New
- Research Article
- 10.1038/s41420-025-02781-6
- Nov 7, 2025
- Cell death discovery
- Raniya Khadiullina + 8 more
The TP53 gene is one of the most frequently mutated genes in human cancers. Mutations often result in loss of tumor-suppressive functions and acquisition of oncogenic properties by p53, contributing to tumor progression and resistance to therapy. Among structural p53 mutations, Y220C is particularly notable for creating a surface-exposed hydrophobic pocket that destabilizes the protein while preserving partial function, making it a promising target for pharmacological reactivation. In this study, we performed a structure-guided phenotypic screen of an in-house heterocyclic compound library to identify novel small-molecule modulators of p53-Y220C. This led to the identification of a series of (1H-pyrrol-1-yl)indazole derivatives (JC16, JC36, JC65), structurally inspired by known Y220C binders. JC16 and JC36 exhibited selective cytotoxicity and pro-apoptotic activity in p53-Y220C mutant cancer cell lines, with minimal effects in wild-type or p53-null cells. These compounds induced a mutant-to-wild-type conformational shift in cellular p53-Y220C, accompanied by transcriptional activation of canonical p53 target genes, including BBC3 (PUMA) and MDM2. Western blot analysis revealed that in HUH7 cells, these effects occurred without a corresponding increase in total p53 protein levels, suggesting a mechanism based on conformational reactivation. Our findings position JC16 and JC36 as early-stage chemical leads with potential to restore mutant p53 function in a context-dependent manner. While their exact mechanism of action remains to be fully elucidated, these results provide a foundation for further development of indazole-based scaffolds as reactivators of the p53-Y220C mutant in cancer therapy.
- New
- Research Article
- 10.1002/adhm.202504291
- Nov 7, 2025
- Advanced healthcare materials
- Liqiao Chen + 13 more
The presence of microplastics in the human body has been widely confirmed. This study aimed to characterize the presence of microplastics in human gastric tissue and explore the relationship between microplastic exposure and gastric cancer risk, as well as the potential underlying molecular mechanisms. Microplastics present in human gastric tissues are characterized using three methods. Following this, the relationship between microplastic exposure and the development of gastric cancer, as well as the potential underlying molecular mechanisms, is explored using clinical case data and transcriptome sequencing. Microplastics are widely present in human gastric tissues, with a significantly higher microplastic load observed in gastric tumor tissues than para-tumor and normal gastric tissues. Furthermore, a high microplastic load is significantly associated with lymph node metastasis in patients with gastric cancer. A high microplastic load leads to significant enrichment of biological processes and signaling pathways, such as cytotoxicity and oxidative stress. Additionally, a high microplastic load is associated with significant upregulation of genes associated with malignant disease, which is significantly associated with poor prognosis in patients with gastric cancer. Microplastic exposure may be a potential risk factor for the onset of gastric cancer.
- New
- Research Article
- 10.1021/acs.jproteome.5c00612
- Nov 7, 2025
- Journal of proteome research
- Judith Nwaiwu + 10 more
Breast cancer is the second most common cause of brain metastasis, often in advanced-stage disease. The mechanisms underlying breast cancer brain metastasis (BCBM), particularly how tumor cells cross the blood-brain barrier and adapt to the brain environment, remain unclear. Cell surface glycosylation plays diverse roles, and its dysregulation in cancer can disrupt signaling and promote metastasis. We investigated how changes in cell surface N-glycans contribute to BCBM by analyzing N-glycans released from human breast cancer cell lines (MDA-MB-231, MDA-MB-361, HTB-131, HTB-22), a brain-seeking variant (MDA-MB-231BR), and glioblastoma cells (CRL-1620) using nano liquid chromatography-tandem mass spectrometry (LC-MS/MS). Surface N-glycans were enzymatically released from live cells with PNGase F without compromising the membrane integrity. Results showed the 231BR cell line expressed higher levels of sialylated N-glycans than other cells, with N-glycan 4502 being the most abundant. Four sialylated structures (4501, 4502, 3501, and 5602) were significantly elevated in 231BR, suggesting a role in brain metastasis. This study expands our understanding of altered N-glycan profiles in BCBM and highlights potential molecular features linked to brain colonization. Further research on these N-glycans could clarify their function in mediating metastasis and identifying therapeutic targets.
- New
- Research Article
- 10.1038/s41467-025-64821-0
- Nov 6, 2025
- Nature communications
- Shigenari Nukaga + 30 more
RNA splicing dysregulation has emerged as a hallmark of cancer and a promising therapeutic target; however, its full landscape in human solid cancer remains poorly characterized. To address this, we perform alternative splicing analyses using RNA-sequencing data from 751 lung adenocarcinoma samples from our cohort integrated with 519 samples from The Cancer Genome Atlas. Visualization of splicing patterns using t-distributed stochastic neighbor embedding reveals substantial inter-tumor heterogeneity driven by distinct molecular subtypes and histological differentiation. We identify a unique molecular subtype associated with inactivating mutations in CMTR2, which encodes Cap-specific mRNA (nucleoside-2'-O-)-methyltransferase 2. CMTR2 mutations are observed in 3.8% of cases and are predominantly truncating mutations, which form an isolated cluster within the splicing landscape. Intrinsic and CRISPR-Cas9-engineered CMTR2 mutations disrupt alternative splicing and sensitize cancer cells to sulfonamide-based RNA splicing modulators and immune checkpoint blockade therapy. Retrospective patient data confirm the increased sensitivity of CMTR2-deficient tumors to immune checkpoint blockade therapy. These findings uncover a previously unrecognized RNA splicing deficiency in human cancers and define a molecular subtype of lung adenocarcinoma driven by RNA splicing dysregulation, suggesting targets for therapeutic intervention in lung cancer.
- New
- Research Article
- 10.12982/cmjs.2025.095
- Nov 6, 2025
- Chiang Mai Journal of Science
- Kyaw Htet Hein + 6 more
Schizophyllum commune, commonly known as the split gill mushroom, is a well-known source of beneficial bioactive compounds that support human health. However, potential uses of peptides generated by digestion of the protein extract from this mushroom has not been well studied. In this study, the proteins extracted from S. commune were isolated and subsequently hydrolyzed with Alcalase® (80 mU/g of protein powder) to produce S. commune protein hydrolysates. Fractionation of the hydrolysate was performed by ultrafiltration with 50, 10, and 3 kDa molecular weight (MW) cut-off. The biological activities of the protein hydrolysates including their anti-cancer and antioxidant properties were found to be different from those of the non-hydrolyzed proteins. The antioxidant activity of protein hydrolysates exhibited the greatest 50% inhibitory concentration (IC50) values as assessed by DPPH and ABTS radical scavenging assay. The effectiveness of the hydrolysate in reducing the survival rate of the SW480 human colorectal cancer cell line was also examined. The protein hydrolysates markedly reduce the viability of the SW480 cell line in comparison to the crude intact protein. These findings suggested that protein hydrolysates obtained from the fruiting body of S. commune had potential applications as a functional food additive.
- New
- Research Article
- 10.1158/2326-6066.cir-25-0343
- Nov 6, 2025
- Cancer immunology research
- Abir K Panda + 18 more
Immune check-point blockade for the treatment of malignancies has been focused on reversing inhibitory pathways in T lymphocytes. Natural killer (NK) cells are a potent innate defense against tumors and virally infected cells, but their therapeutic manipulation for anti-cancer immunity has been inadequately explored. Considerable attention has been focused on approaches to blocking inhibitory receptors on NK and myeloid cells. Most effort has been directed to the killer immunoglobulin-like receptors (KIR) and CD94/NKG2A on NK cells. Another set of receptors with similar function in both NK cells and myeloid cells is the leukocyte immunoglobulin like receptors (LILR) that interact with a wide variety of HLA molecules. Using pan-anti-HLA mAbs that recognize a conserved epitopic region on HLA also seen by LILR, we investigated their functional effects in several models of tumor immunity. The pan-anti-HLA-mAbs blocked the binding of most LILRs, did not block killer cell immunoglobulin-like receptors (KIR) or CD94/NKG2A/C or TCR recognition. They also activated dysfunctional NK cells explanted from a variety of human cancers, and resulted in enhancement of tumor immunity in humanized mice. The mAbs also exert direct anti-tumor effects. These results suggest that activation of innate immunity via disruption of HLA/LILR interactions is a potent approach for control of both primary tumors and potentially tumor metastases.
- New
- Research Article
- 10.3390/cancers17213571
- Nov 5, 2025
- Cancers
- Zhihai Deng + 11 more
Background: Prostate cancer, an epithelial malignancy occurring in the prostate, is the most common malignant tumor of the male genitourinary system and has a low survival rate in advanced prostate cancer after metastasis. It is urgent to explore novel therapeutic targets and strategies for treating prostate cancer. Circular RNA (circRNA) and ferroptosis both play critical roles in prostate cancer progression. However, the regulatory effect of circRNA on ferroptosis remains unclear. Methods: Here, circRNA expression profiles in prostate cancer were explored by bioinformatics analysis and human prostate cancer tissue microarray. Stable circRNA-knockdown or overexpressed prostate cancer cell lines were constructed by lentivirus. AGO2-RNA immunoprecipitation (AGO2-RIP) was utilized to identify circRNA-microRNA (miRNA) interaction. Results: Results of this study indicate that circATP2C1 is highly expressed in prostate cancer. In addition, circATP2C1 promotes prostate cancer cell proliferation, migration, and invasion by suppressing ferroptosis in vitro. Moreover, circATP2C1 facilitates the tumorigenicity of prostate cancer by inhibiting ferroptosis in vivo. Conclusions: Mechanistically, circATP2C1 hinders ferroptosis by increasing solute carrier family 7 member 11 (SLC7A11) expression via sponging miR-654-3p. In summary, our findings highlight the oncogenic role of circATP2C1 in prostate cancer and provide novel targets and strategies for treating prostate cancer.
- New
- Research Article
- 10.3390/molecules30214310
- Nov 5, 2025
- Molecules
- Ifeoma Roseline Ezeanolue + 5 more
This scoping review synthesized evidence from 2015 to 2025 on the anti-inflammatory and anticancer potential of pecan (Carya illinoinensis) kernel extracts, focusing on bioactive composition and cell signaling pathway modulation. Pecan kernels contain diverse phenolic compounds including gallic acid, catechin, epicatechin, and ellagic acid, along with tocopherols and unsaturated fatty acids, exhibiting significant cultivar-dependent variation influenced by ripening stage, processing conditions, and orchard management practices. In vitro studies demonstrate that kernel extracts possess substantial antioxidant capacity and exert antiproliferative and cytotoxic effects against various human cancer cell lines, including colon cancer cells, with evidence of apoptosis induction. Extraction methodologies significantly influence bioactive compound recovery and biological activity, with both lipid and phenolic fractions contributing to therapeutic potential. While current evidence highlights promising anti-inflammatory and anticancer properties mediated through modulation of apoptotic pathways, research remains predominantly limited to compositional analyses and in vitro models. Future investigations should elucidate specific molecular mechanisms, identify precise signaling pathway targets, conduct in vivo validation studies, and optimize processing conditions to maximize bioactive retention for potential therapeutic applications in cancer prevention and treatment.
- New
- Research Article
- 10.1038/s41598-025-19296-w
- Nov 5, 2025
- Scientific reports
- Rajashree Panigrahi + 3 more
Kaempferia parviflora (black ginger) is a tropical edible medicinal plant with valuable rhizome product used in the pharmaceutical industry for wide beneficial health effects in curing various diseases. The current study aims to analyse compounds present in rhizome oil using Gas Chromatography-Time of Flight-Mass Spectrometry (GC-TOF-MS). Hydro distillation was used to extract the essential oil from the plant rhizomes. 129 compounds were identified by GC TOF-MS analysis, with the main components being benzenepropanol, à-methyl-acetate (9.78 ± 0.01%), camphene (9.39 ± 0.01%), α-pinene (7.50 ± 0.01%), linalool (5.46 ± 0.01%), and camphore (5.04 ± 0.01%). Moreover, in this investigation, Benzenepropanol, à-methyl-, acetate, is Reported for the first time as a major constituent. The cytotoxic potential of the oil was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against a broad spectrum of human cancer cell lines, including the IC50 value of A549 (38.73 ± 0.01µg/mL), A431 (68.33 ± 0.02µg/mL), HCT-15 (68.33 ± 0.01µg/mL), KB (88.57 ± 0.03µg/mL), LN-18 (43.23 ± 0.01µg/mL), Panc-1 (46.68 ± 0.01µg/mL), and SKBR-3(11.79 ± 0.01µg/mL). HEK293 (non-cancer cell line) was also included to check its cytotoxicity towards normal cells. Antimicrobial activities were assessed against Staphylococcus aureus, Bacillus subtilis, Staphylococcus epidermidis, Klebsiella pneumoniae and Candida albicans using disc diffusion method, showing highest zone of inhibition (ZOI) against Klebsiella pneumoniae, Candida albicans (30 ± 0.01mm) and the lowest against Bacillus subtilis (16 ± 0.02mm). The antioxidant activity revealed better IC50 values in both 2,2-diphenyl-1-picrylhydrazyl (DPPH) as 0.50 ± 0.01mg/ml and 2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) as 0.40 ± 0.01mg/ml assays. Hence, the obtained cytotoxic, antimicrobial and antioxidant potential of the tested oil, proved the potent quality of the Kaempferia parviflora oil. Hence, it can be used as an alternative antibiotic as well as antioxidant agent in the field of medicine and its commercialization in the global market.
- New
- Research Article
- 10.1186/s12935-025-04032-1
- Nov 5, 2025
- Cancer cell international
- Abbas Asoudeh-Fard + 2 more
Probiotics, particularly Lactobacillus species, show promise as adjuvants in cancer therapy due to their pro-apoptotic effects. This study investigated the synergistic impact of Lactobacillus fermentum (Ab.RS23) and vincristine sulfate on colorectal (HT-29) and breast (MCF-7) cancer cells. Cells were treated with vincristine, L. fermentum, or both. Cell viability was measured by MTT assay. Apoptosis was analyzed via Annexin V-FITC/PI flow cytometry. Gene expression changes were evaluated by RT-qPCR. Co-treatment reduced the IC₅₀ of vincristine by 8-fold in HT-29 and 13-fold in MCF-7 cells. Apoptotic signaling was enhanced, with pro-apoptotic pathways upregulated and survival pathways downregulated. L. fermentum enhanced vincristine-induced apoptosis and reduced the required drug dose, which may contribute to lowering vincristine-associated toxicity. These findings require confirmation through in vivo studies.
- New
- Research Article
- 10.3390/cells14211736
- Nov 5, 2025
- Cells
- Ildikó Vig + 6 more
The human BUD31 gene has been associated with various processes including cancer. To better understand its function, we used genetic methods to study Schizosaccharomyces pombe cells lacking the BUD31 homologous gene (cwf14) and performed sequence analysis using bioinformatics methods. Mutant cells lacking the cwf14 gene showed cell size and division defects, altered stress response, rapamycin sensitivity, enhanced chronological aging, and increased sporulation tendency. These processes are known to be regulated by the TOR pathway. The cwf14-TOR link was also supported by further experiments. We demonstrated that most protein-coding genes affected by cwf14 deletion are upregulated, encode hydrolases, oxidoreductases, and are often involved in transport. GO enrichment drew our attention to genes related to nitrogen transport, while additional data pointed to a nutrient/nitrogen (N) sensing problem. Although Cwf14 protein is associated with spliceosome complex, most genes affected by the absence of cwf14 do not contain introns, suggesting that they are influenced indirectly by the cwf14 gene. In silico experiments have revealed that BUD31 orthologous genes are found from yeast to humans, are evolutionarily conserved with a high degree of sequence identity, conserved motifs, and structures. Since the human gene partially complemented the mutant phenotype of S. pombe cells, indicating functional homology, our data can help better understand pathological mechanisms observed in human cancer cells.
- New
- Research Article
- 10.3390/pharmaceutics17111433
- Nov 5, 2025
- Pharmaceutics
- Kateřina Běhalová + 5 more
Background/Objectives: Here, we report the design, synthesis, and in vitro biological evaluation of a novel stimuli-sensitive nanotherapeutics based on cisplatin analog, cis-[PtCl2(NH3)(2-(3-oxobutyl)pyridine)] (Pt-OBP), covalently linked to a N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer via a pH-sensitive hydrazone bond. Methods: Two polymer–drug conjugates, P-Pt-A and P-Pt-B, were synthesized, differing in spacer length between the polymer chain and hydrazone bond, which in turn modulates their drug release kinetics. Results: The spacer based on hydrazone bond demonstrated satisfactory stability under blood-mimicking conditions while enabling selective release of the active drug intracellularly or even in the mildly acidic tumor microenvironment. Pt-OBP exhibits comparable or even superior cytostatic and cytotoxic activity to carboplatin across a panel of murine and human cancer cell lines, with the highest potency observed in FaDu cells representing human head and neck squamous cell carcinoma. Mechanistically, Pt-OBP induced significant phosphorylation of γ-H2AX and activation of caspase-3, indicating its ability to cause DNA damage with subsequent apoptosis induction. P-Pt-A retained moderate biological activity, whereas the slower-releasing P-Pt-B exhibited reduced potency in vitro, consistent with its drug release profile. Conclusions: Notably, free Pt-OBP induced rapid apoptotic cell death, surpassing carboplatin at early time points, and the polymeric conjugates achieved comparable pro-apoptotic activity after extended incubation, suggesting effective intracellular release of the active drug.
- New
- Research Article
- 10.1038/s43587-025-00984-1
- Nov 4, 2025
- Nature aging
- Adrienne Parsons + 12 more
Aging is associated with increased breast cancer risk, and the oldest and youngest patients have worse outcomes, irrespective of subtype. It is unknown how age affects cells in the breast tumor microenvironment or how they contribute to age-related pathology. Here we discover age-associated differences in cell states in human estrogen receptor-positive and triple-negative breast cancers using analyses of existing bulk and single-cell transcriptomic data. We generate and apply an Age-Specific Program ENrichment (ASPEN) analysis pipeline, revealing age-related changes, including increased tumor cell epithelial-mesenchymal transition and cancer-associated fibroblast inflammatory responses in triple-negative breast cancer. Estrogen receptor-positive breast cancer displays increased ESR1 expression and reduced vascular and immune cell metabolism with age. Cell interactome analysis reveals candidate signaling pathways that drive age-related cell states. Spatial analyses across independent clinical cohorts support the computational findings. This work identifies potential targets for age-adapted therapeutic interventions for breast cancer.
- New
- Research Article
- 10.1038/s41598-025-22389-1
- Nov 4, 2025
- Scientific Reports
- Chiaki Yagi + 17 more
Oncolytic adenoviruses replicate selectively in tumor cells and induce immunogenic cell death, but predictive biomarkers for early therapeutic response are lacking. This study evaluated extracellular vesicle-encapsulated adenoviral E1A-DNA (EV-E1A-DNA) as a minimally invasive biomarker for monitoring responses to telomerase-specific oncolytic adenoviruses OBP-301 and OBP-502. EVs were isolated from human and murine cancer cell lines and from the serum of treated mice using ultracentrifugation. EV-associated E1A-DNA levels were measured by quantitative polymerase chain reaction and found to correlate with cytotoxicity in vitro and tumor regression in vivo. In xenograft models, serum EV-E1A-DNA levels at 2 days post-treatment showed strong correlations with final tumor volume and survival, supporting their utility as an early predictive biomarker. In immunocompetent mice pre-immunized with wild-type adenovirus, free viral DNA was undetectable in serum due to neutralizing antibodies, whereas EV-E1A-DNA remained detectable. This “stealth effect” indicates that EVs protect viral components from immune clearance. These results demonstrate that EV-E1A-DNA is a sensitive and virus-specific biomarker that enables early assessment of therapeutic efficacy, even in the presence of antiviral immunity. This strategy offers a promising liquid biopsy approach for personalized monitoring of oncolytic virotherapy and may be applicable to other virus-based therapies.Supplementary InformationThe online version contains supplementary material available at 10.1038/s41598-025-22389-1.
- New
- Research Article
- 10.1038/s43587-025-00986-z
- Nov 4, 2025
- Nature aging
- Emily G Shuldiner + 14 more
Most cancers are diagnosed in people over 60 years of age, but little is known about how age impacts tumorigenesis. While aging is accompanied by mutation accumulation (widely understood to contribute to cancer risk) it is associated with numerous other cellular and molecular changes likely to impact tumorigenesis. Moreover, cancer incidence decreases in the oldest part of the population, suggesting that very old age may reduce carcinogenesis. Here we show that aging represses oncogenic KRAS-driven tumor initiation and growth in genetically engineered mouse models of human lung cancer. Moreover, aging dampens the impact of inactivating many tumor suppressor genes with the impact of inactivating PTEN, a negative regulator of the PI3K-AKT pathway, weakened disproportionately. Single-cell transcriptomic analysis revealed that neoplastic cells in aged mice retain age-related transcriptomic changes, showing that the impact of age persists through oncogenic transformation. Furthermore, the consequences of PTEN inactivation were strikingly age-dependent, with PTEN deficiency reducing signatures of aging in cancer cells and the tumor microenvironment. Our findings underscore the interconnectedness of the pathways involved in aging and tumorigenesis and document tumor-suppressive effects of aging that may contribute to the deceleration in cancer incidence with age.
- New
- Research Article
- 10.1021/acsnano.5c10776
- Nov 4, 2025
- ACS nano
- Shuang Chang + 15 more
Cancer-associated fibroblasts (CAFs) are heterogeneous and critical drivers of tumor progression, yet engineering tumor-promoting CAF subtypes in situ offers an untapped therapeutic opportunity. Herein, we verify that the FAP+αSMA+ CAFs subtype in the 4T1 murine model effectively recapitulates its counterpart in human triple-negative breast cancer (TNBC), demonstrating strong tumor-promoting activity. We specifically engineer these CAFs in situ to enhance antitumor immunity using an innovative nanodrug, IL-15 plasmid-loaded FAP-sensitive MgCa/z-Gly-Pro-pamidronate acid nanoparticles (PN/MCG NPs). PN/MCG NPs can reverse the tumor-promoting phenotype of FAP+ αSMA+ CAFs and engineer FAP+ αSMA+ CAFs to sustain IL-15 expression. These engineered FAP+ αSMA+ CAFs significantly reduce the tumor immune suppression. Gene set enrichment analysis (GSEA) reveals enhanced immune cell proliferation and activation. Furthermore, we also prove that FAP+ αSMA+ human mammary fibroblast cells (FAP+ αSMA+ HMFs) also can be engineered by PN/MCG NPs in vitro. Our findings demonstrate that in situ CAF engineering is a promising strategy to remodel the tumor microenvironment and enhance immunotherapy in TNBC.
- New
- Research Article
- 10.7717/peerj.20316
- Nov 4, 2025
- PeerJ
- Ahmed Abdullah Al Awadh + 3 more
Background Ovarian cancer is a lethal gynecological malignancy, largely due to late-stage diagnosis and poor prognosis. MicroRNA-597-3p (miR-597-3p) has been identified as a tumor suppressor in several cancers, while metastasis-associated colon cancer 1 (MACC1) functions as an oncogene that promotes metastasis. This study investigated the role of miR-597-3p and its regulation of MACC1 in ovarian cancer progression. Methods Ovarian cancer cell lines and the normal ovarian epithelial cell line IOSE-398 were used. Quantitative real-time PCR (qRT-PCR) measured the expression of miR-597-3p and MACC1. Functional assays (MTT, colony formation, AO/EB staining, and Transwell invasion) evaluated cell proliferation, cell death, and invasion. Dual-luciferase reporter assays confirmed the interaction between miR-597-3p and MACC1, while bioinformatics analysis identified potential targets. Western blotting was used to validate MACC1 and downstream proteins, MMP-2, and MMP-9 at the protein level. Results MiR-597-3p was significantly downregulated ( P < 0.05) in ovarian cancer cells, whereas MACC1 was upregulated. Overexpression of miR-597-3p suppressed cell viability, colony formation, and invasion, and increased cell death with a pro-apoptotic shift in Bax/Bcl-2 expression. Mechanistically, miR-597-3p directly targeted MACC1, leading to reduced expression of MMP-2 and MMP-9 at both transcript and protein levels. Notably, MACC1 overexpression reversed the tumor-suppressive effects of miR-597-3p. Conclusions MiR-597-3p functions as a tumor suppressor in ovarian cancer by directly targeting MACC1, thereby inhibiting proliferation, invasion, and survival. These findings highlight the miR-597-3p/MACC1 axis as a potential therapeutic target and suggest miR-597-3p as a promising biomarker for ovarian cancer.
- New
- Research Article
- 10.1371/journal.pone.0335934.r008
- Nov 3, 2025
- PLOS One
- Md Sharif Hasan + 10 more
HMGA1, an architectural transcription factor that plays a crucial role in tumorigenesis, chemotherapy resistance and cancer stem cell transformation in many human cancers, is intrinsically disordered and cannot be targeted by conventional small molecule drug therapy. While HMGA1 is required and essential for normal growth and development, HMGA1 expression occurs at very low levels in normal healthy adult cells. In contrast, HMGA1 is expressed at very high levels in many different types of human cancer cells. Since HMGA1 cannot be targeted using conventional small molecule drug therapy, alternative approaches are needed to target HMGA1 in new cancer therapies. Here, we explored the use of serotype 5 adenoviruses (Ad5) engineered 1) to sequester overexpressed HMGA1 in cancer cells using an HMGA1 hyper binding site (HBS) inserted into the Ad5 genome and 2) to suppress HMGA1 synthesis in cancer cells by incorporating exogenous genes into the Ad5 genome that encode an artificial HMGA1 cis-antisense transcript (AAT) and that encode a gene to express an HMGA1-targeted shRNA transcript (shRNA). The three engineered Ad5s were tested in MiaPaCa-2, PANC-1 and BxPC-3 human pancreatic cancer cell lines and in the ZR-75 human breast cancer cell line. Cancer cell viabilities and cell migration capability decreased by ~50–75% with HBS viruses and by 25–50% for shRNA and AAT viruses. Anchorage-independent migration capacity decreased by 60–70% with all three HBS, shRNA and AAT viruses. HMGA1 mRNA transcripts levels varied from 100 to 300 copies per cell in untreated cells and these levels were not significantly affected by treatment with the HBS and shRNA viruses, however the HMGA1 mRNA levels increased by ~3-fold upon AAT virus treatment. HMGA1 protein levels decreased in the range of 40, 50 and 70% with shRNA, AAT and HBS viruses, respectively. The HBS virus designed to sequester HMGA1 proved most effective overall in suppressing HMGA1 oncogenic activity in these in vitro cell-based studies compared to the AAT and shRNA viruses.
- New
- Research Article
- 10.1371/journal.pone.0335934
- Nov 3, 2025
- PloS one
- Md Sharif Hasan + 7 more
HMGA1, an architectural transcription factor that plays a crucial role in tumorigenesis, chemotherapy resistance and cancer stem cell transformation in many human cancers, is intrinsically disordered and cannot be targeted by conventional small molecule drug therapy. While HMGA1 is required and essential for normal growth and development, HMGA1 expression occurs at very low levels in normal healthy adult cells. In contrast, HMGA1 is expressed at very high levels in many different types of human cancer cells. Since HMGA1 cannot be targeted using conventional small molecule drug therapy, alternative approaches are needed to target HMGA1 in new cancer therapies. Here, we explored the use of serotype 5 adenoviruses (Ad5) engineered 1) to sequester overexpressed HMGA1 in cancer cells using an HMGA1 hyper binding site (HBS) inserted into the Ad5 genome and 2) to suppress HMGA1 synthesis in cancer cells by incorporating exogenous genes into the Ad5 genome that encode an artificial HMGA1 cis-antisense transcript (AAT) and that encode a gene to express an HMGA1-targeted shRNA transcript (shRNA). The three engineered Ad5s were tested in MiaPaCa-2, PANC-1 and BxPC-3 human pancreatic cancer cell lines and in the ZR-75 human breast cancer cell line. Cancer cell viabilities and cell migration capability decreased by ~50-75% with HBS viruses and by 25-50% for shRNA and AAT viruses. Anchorage-independent migration capacity decreased by 60-70% with all three HBS, shRNA and AAT viruses. HMGA1 mRNA transcripts levels varied from 100 to 300 copies per cell in untreated cells and these levels were not significantly affected by treatment with the HBS and shRNA viruses, however the HMGA1 mRNA levels increased by ~3-fold upon AAT virus treatment. HMGA1 protein levels decreased in the range of 40, 50 and 70% with shRNA, AAT and HBS viruses, respectively. The HBS virus designed to sequester HMGA1 proved most effective overall in suppressing HMGA1 oncogenic activity in these in vitro cell-based studies compared to the AAT and shRNA viruses.