Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (468)

Search Parameters:
Keywords = E6/E7 oncogene expression

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 3196 KB  
Article
Development of RALA-Based Mannosylated Nanocarriers for Targeted Delivery of Minicircle DNA Vaccines Encoding HPV-16 Oncogenes
by Andressa Giusti, Dalinda Eusébio, Matilde Costa, Inês Silveira, Swati Biswas, Diana Costa and Ângela Sousa
Vaccines 2026, 14(1), 18; https://doi.org/10.3390/vaccines14010018 - 23 Dec 2025
Viewed by 291
Abstract
Background/Objectives: Cervical cancer is a leading cause of cancer-related mortality among women, primarily driven by persistent infections with high-risk human papillomavirus (HPV), particularly HPV-16. Vaccines based on plasmid DNA encoding the viral oncogenes E6 and E7 represent a promising immunotherapeutic strategy, but their [...] Read more.
Background/Objectives: Cervical cancer is a leading cause of cancer-related mortality among women, primarily driven by persistent infections with high-risk human papillomavirus (HPV), particularly HPV-16. Vaccines based on plasmid DNA encoding the viral oncogenes E6 and E7 represent a promising immunotherapeutic strategy, but their efficacy remains limited due to poor cellular uptake. Cell-penetrating peptides such as RALA improve intracellular delivery, and functionalization with octa-arginine peptide conjugated to mannose (R8M) further enhances targeting of antigen-presenting cells (APCs). This study aimed to obtain the minicircle DNA (mcDNA) encoding mutant HPV-16 E6 and/or E7 antigens, and optimize its complexation with mannosylated RALA-based nanoparticles to improve vector delivery and consequently antigen presentation. Methods: Nanoparticles were formulated at different concentrations of RALA, with and without R8M functionalization. Their characterization included hydrodynamic diameter, polydispersity index, zeta potential, complexation efficiency (CE), stability, morphology, and Fourier-Transform Infrared Spectroscopy. In vitro assays in JAWS II dendritic cells (DCs) assessed biocompatibility, transfection efficiency and target gene expression. Results: Optimal conditions were obtained at 72.5 µg/mL of RALA, producing nanoparticles smaller than 150 nm with high CE (>97%) and uniform size distribution. Functionalization with R8M at 58 µg/mL preserved these characteristics when complexed with all mcDNA vectors. The formulations were biocompatible and effectively transfected DCs. Mannosylated formulations enhanced antigenic expression compared to non-mannosylated counterparts, evidencing a mannose-receptor-mediated uptake, while increasing the production of pro-inflammatory cytokines. Conclusions: Nanoparticles based on the RALA peptide and functionalized with R8M significantly improved mcDNA transfection and gene expression in APCs. These findings support further investigation of this system as a targeted DNA vector delivery platform against HPV-16. Full article
(This article belongs to the Special Issue New Approaches to Vaccine Development and Delivery)
Show Figures

Figure 1

16 pages, 677 KB  
Review
MDM2 in Tumor Biology and Cancer Therapy: A Review of Current Clinical Trials
by Francesco Russano, Mattia Sturlese, Luigi Dall’Olmo, Francesco Callegarin, Davide Brugnolo, Paolo Del Fiore, Vittoria Patti, Arianna Purpura, Stefano Moro, Marco Rastrelli and Simone Mocellin
Int. J. Mol. Sci. 2026, 27(1), 99; https://doi.org/10.3390/ijms27010099 - 22 Dec 2025
Viewed by 241
Abstract
The Murine Double Minute 2 (MDM2) gene encodes an E3 ubiquitin ligase that negatively regulates the tumor suppressor p53, maintaining low p53 levels through ubiquitination and proteasomal degradation. MDM2 overexpression in various malignancies leads to reduced p53 activity, contributing to tumor [...] Read more.
The Murine Double Minute 2 (MDM2) gene encodes an E3 ubiquitin ligase that negatively regulates the tumor suppressor p53, maintaining low p53 levels through ubiquitination and proteasomal degradation. MDM2 overexpression in various malignancies leads to reduced p53 activity, contributing to tumor initiation and resistance to therapies. As such, MDM2 is a promising target for drug development. Innovative small-molecule inhibitors are being designed to disrupt the MDM2-p53 interaction, thereby restoring p53’s tumor-suppressive functions. This review focuses on clinical trials evaluating MDM2 inhibition for cancer therapy. MDM2 exerts its oncogenic effects primarily through its interaction with p53 but also has p53-independent functions involved in cell cycle progression and DNA repair. Elevated MDM2 expression is associated with poor prognosis across various cancers, including dedifferentiated liposarcoma, breast cancer, and glioblastoma. Targeting MDM2 with inhibitors has shown promising potential in clinical development, aiming to reactivate p53’s functions in tumors with wild-type TP53, improving therapeutic outcomes in cancer treatment. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

12 pages, 1469 KB  
Opinion
The Complexity of Bovine Leukemia Virus Oncogenesis
by Florine Doucet, Alexis Fontaine, Malik Hamaidia, Jean-Rock Jacques, Thomas Jouant, Nour Mhaidly, Songkang Qin, Roxane Terres, Xavier Saintmard, Luc Willems and Manon Zwaenepoel
Viruses 2025, 17(12), 1609; https://doi.org/10.3390/v17121609 - 12 Dec 2025
Viewed by 273
Abstract
Bovine leukemia virus (BLV) is a retrovirus infecting several bovid species, notably Bos taurus, where it fulfills Koch’s postulates for pathogenicity. The virus primarily targets B-lymphocytes, establishing lifelong infections that remain mostly asymptomatic but can progress to lymphocytosis or lymphoma. Transmission occurs [...] Read more.
Bovine leukemia virus (BLV) is a retrovirus infecting several bovid species, notably Bos taurus, where it fulfills Koch’s postulates for pathogenicity. The virus primarily targets B-lymphocytes, establishing lifelong infections that remain mostly asymptomatic but can progress to lymphocytosis or lymphoma. Transmission occurs through live infected cells via blood, milk, or transplacental routes. Despite a robust antiviral immunity, BLV replicates by producing virions (i.e., the infectious cycle) or inducing mitosis of infected cells (i.e., clonal expansion). The immune system effectively controls the infectious cycle but fails to impede clonal expansion, leading to chronic immune activation and immunosuppression. BLV modifies the transcriptome of the host cell by expressing oncogenic factors (Tax), viral microRNAs and antisense RNAs. Leukemogenesis arises from cumulative alterations of the virus (e.g., 5′-end deletions of the integrated provirus and histone modifications of the LTR promoter) and the host cell (e.g., genomic mutations and favorable chromatin integration). This model underscores a unique persistence strategy, linking chronic infection, immune evasion, and slow multistep oncogenesis in the bovine host. Full article
Show Figures

Graphical abstract

30 pages, 1506 KB  
Review
Lactate-Mediated Epigenetic and Immunometabolic Reprogramming in Glioma: An Emerging Axis Linking Metabolism to Tumor Progression
by Xinyi Xie, Wenjing Zhou, Yin Ku, Shasha Li, Yunhao Yang, Xiaohu Hao and Yaohui Chen
Biomedicines 2025, 13(12), 3041; https://doi.org/10.3390/biomedicines13123041 - 11 Dec 2025
Viewed by 587
Abstract
Background: Among primary malignant brain tumors in adults, glioblastoma is the most common and the most aggressive, characterized by profound metabolic reprogramming. This metabolic shift is essential for sustaining relentless proliferation and adapting to the challenging tumor microenvironment (TME). Central to this [...] Read more.
Background: Among primary malignant brain tumors in adults, glioblastoma is the most common and the most aggressive, characterized by profound metabolic reprogramming. This metabolic shift is essential for sustaining relentless proliferation and adapting to the challenging tumor microenvironment (TME). Central to this adaptation in glioma is the Warburg effect, which leads to excessive lactate production and accumulation, even in the presence of oxygen. This metabolic divergence significantly impacts the tumor immune microenvironment, promoting the recruitment of immunosuppressive cells and weakening the anti-tumor immune response. Core Content: This review provides a comprehensive analysis of the multifaceted roles of lactate in IDHwt glioma pathogenesis. It explores how lactate serves as a critical nexus connecting aberrant metabolism, epigenetic reprogramming (notably via histone lactylation), and immune evasion. The review delves into the molecular mechanisms by which lactate, particularly through the post-translational modification known as lactylation, directly modulates the epigenetic landscape to promote oncogene expression. Furthermore, it examines lactate’s role in acidifying the TME, promoting the immunosuppressive M2 polarization of glioma-associated macrophages (GAMs), and inhibiting the cytotoxic activity of T lymphocytes. Conclusions: This “lactate-centric” framework provides a unifying model that links metabolic dysregulation directly to malignant progression and therapeutic resistance (e.g., to TMZ). By elucidating this metabolic–epigenetic–immune axis, the review highlights a critical dependency that fuels glioma aggression. Finally, it discusses emerging therapeutic strategies aimed at targeting lactate production (LDHAi), transport (MCTi), and downstream epigenetic signaling (HDACi/p300i), offering novel avenues for integrative immunometabolic therapy. Full article
(This article belongs to the Special Issue Diagnosis, Pathogenesis and Treatment of CNS Tumors (2nd Edition))
Show Figures

Figure 1

22 pages, 2808 KB  
Article
Utility of Tumor Suppressor E2F Target Gene Promoter Elements to Drive Gene Expression Specifically in Cancer Cells
by Kenta Kurayoshi, Masakazu Tanaka, Rinka Nakajima, Yaxuan Zhou, Mashiro Shirasawa, Mariana Fikriyanti, Jun-ichi Fujisawa, Ritsuko Iwanaga, Andrew P. Bradford, Keigo Araki and Kiyoshi Ohtani
Cells 2025, 14(24), 1953; https://doi.org/10.3390/cells14241953 - 9 Dec 2025
Viewed by 320
Abstract
The transcription factor E2F is the principal target of the tumor suppressor pRB. In almost all cancers, pRB function is disabled due to oncogenic changes, leading to enhanced E2F activity, thereby facilitating aberrant cell proliferation. Enhanced E2F activity has been utilized to drive [...] Read more.
The transcription factor E2F is the principal target of the tumor suppressor pRB. In almost all cancers, pRB function is disabled due to oncogenic changes, leading to enhanced E2F activity, thereby facilitating aberrant cell proliferation. Enhanced E2F activity has been utilized to drive gene expression preferentially in cancer cells using E2F target promoters, such as the E2F1 promoter. However, these promoters are also activated by physiological E2F activity in normal proliferating cells, resulting in gene expression in normal proliferating cells. In contrast, promoters of tumor suppressor genes, such ARF and TAp73, are activated by deregulated E2F activity, induced by loss of pRB control, but not by physiological E2F activity, induced by growth stimulation, thereby providing a mechanism to drive expression specifically in cancer cells. Here we show artificial promoters, in which E2F-responsive elements of the TAp73 gene are tandemly connected to the ARF core promoter, exhibited higher cancer cell specificity than E2F1, hTERT, or ARF promoters. Moreover, adenoviruses driving a cytotoxic gene using these artificial promoters showed cancer cell-specific cytotoxicity and inhibited tumor growth in a xenograft mouse model. These results indicate utility of tumor suppressor gene promoter elements to drive gene expression specifically in cancer cells. Full article
(This article belongs to the Special Issue Cancer Cell Signaling, Autophagy and Tumorigenesis)
Show Figures

Figure 1

11 pages, 1243 KB  
Article
An ETS2 Enhancer Variant May Modulate Gene Expression and Contribute to Defining a Genetic Risk Profile for SLE Susceptibility
by Andrea Latini, Giada De Benedittis, Chiara Morgante, Carlo Perricone, Fulvia Ceccarelli, Fabrizio Conti, Giuseppe Novelli, Cinzia Ciccacci and Paola Borgiani
Genes 2025, 16(12), 1462; https://doi.org/10.3390/genes16121462 - 8 Dec 2025
Viewed by 264
Abstract
Background/Objectives: Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease strongly influenced by genetic factors. Genome-wide association studies (GWASs) have identified numerous non-coding susceptibility loci, but their functional roles remain poorly understood. The single-nucleotide variant (SNV) rs2836882, located in an enhancer near [...] Read more.
Background/Objectives: Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease strongly influenced by genetic factors. Genome-wide association studies (GWASs) have identified numerous non-coding susceptibility loci, but their functional roles remain poorly understood. The single-nucleotide variant (SNV) rs2836882, located in an enhancer near the ETS2 proto-oncogene, has been implicated in immune regulation, though its contribution to SLE is unclear. Methods: We analyzed rs2836882 in 246 Italian patients with SLE and 216 matched controls using TaqMan genotyping. A weighted genetic risk score (wGRS) combining rs2836882 with other known SLE variants was calculated. ETS2 mRNA expression was quantified by RT-qPCR in PBMCs from 60 individuals, and in silico analyses assessed the variant’s functional context. Results: The rs2836882 risk allele was significantly associated with SLE (OR = 1.54, p = 0.02). Patients showed a markedly higher wGRS than controls (p < 0.00001), confirming an additive genetic burden. In silico data indicated that rs2836882 lies within an active enhancer region (H3K4me1/H3K27ac+) containing PU.1 binding motifs and functions as an expression quantitative trait locus (eQTL) for ETS2. Expression analysis demonstrated that carriers of the risk allele exhibited significantly increased ETS2 expression compared to non-carriers (p = 0.002) in both groups. Conclusions: In conclusion, rs2836882 is a functional regulatory variant that enhances ETS2 transcription and contributes to increased SLE susceptibility. These findings establish a mechanistic link between a non-coding GWAS locus and disease risk, emphasizing the role of regulatory variants in autoimmune pathogenesis and supporting the integration of functional non-coding variants into genetic risk models for improved patient stratification. Full article
(This article belongs to the Special Issue Genetic and Epigenetic Insights in Autoimmune Diseases)
Show Figures

Figure 1

13 pages, 1306 KB  
Review
Plant-Derived miRNAs as Potential Cross-Kingdom Cancer Regulators
by Aizhan Rakhmetullina, Zuzanna Lubas and Piotr Zielenkiewicz
Genes 2025, 16(12), 1441; https://doi.org/10.3390/genes16121441 - 2 Dec 2025
Viewed by 524
Abstract
MicroRNAs (miRNAs) are key posttranscriptional regulators of gene expression that influence cancer initiation, progression, and therapeutic response. While most studies have focused on endogenous miRNAs, emerging evidence has highlighted the role of plant-derived miRNAs as exogenous dietary regulators capable of cross-kingdom gene modulation. [...] Read more.
MicroRNAs (miRNAs) are key posttranscriptional regulators of gene expression that influence cancer initiation, progression, and therapeutic response. While most studies have focused on endogenous miRNAs, emerging evidence has highlighted the role of plant-derived miRNAs as exogenous dietary regulators capable of cross-kingdom gene modulation. This review summarises current knowledge regarding plant-derived miRNAs and their ability to regulate human cancer-related genes. Experimental findings indicate that plant miRNAs can withstand gastrointestinal digestion, enter the circulation, and regulate the expression of oncogenes, tumour suppressors, long noncoding RNAs, and immune checkpoint molecules via canonical RNA-induced silencing mechanisms. Specific examples include miR-156a, miR-159a-3p, miR-166a, miR-167e-5p, miR-171, miR-395e, miR-2911, miR-4995 and miR-5754, which exhibit anticancer activities across various cancer types and modulate key signalling pathways in mammalian cells, highlighting their potential as cross-kingdom regulators with therapeutic relevance. In addition to these characterised miRNAs, certain plant groups, which are rich in bioactive compounds, remain unexplored as sources of functional miRNAs, representing a promising avenue for future research. Collectively, these studies underscore the ability of plant-derived miRNAs to modulate mammalian gene expression and suggest their potential as diet-based or synthetic therapeutic agents. Further investigations into their bioavailability, target specificity, and functional relevance could inform innovative strategies for cancer prevention, integrating nutritional, molecular biological, and therapeutic approaches. Full article
(This article belongs to the Special Issue Function and Regulatory Mechanism of MicroRNAs in Cancers)
Show Figures

Graphical abstract

35 pages, 2185 KB  
Review
Rho Small GTPase Family in Androgen-Regulated Prostate Cancer Progression and Metastasis
by Dontrel William Spencer Hairston, Maria Mudryj and Paramita Mitra Ghosh
Cancers 2025, 17(22), 3680; https://doi.org/10.3390/cancers17223680 - 17 Nov 2025
Viewed by 513
Abstract
Background/Objectives: Rho small GTPases (RSG), which regulates metastasis, constitute eight subfamilies—“classical” Rho, Rac, cdc42, and “atypical” Rif, Rnd, Wrch, RhoH, and RhoBTB. Their downstream signaling requires switching between GTP-bound active and GDP-bound inactive forms. Classical RSGs, but not atypical RSGs, require regulation [...] Read more.
Background/Objectives: Rho small GTPases (RSG), which regulates metastasis, constitute eight subfamilies—“classical” Rho, Rac, cdc42, and “atypical” Rif, Rnd, Wrch, RhoH, and RhoBTB. Their downstream signaling requires switching between GTP-bound active and GDP-bound inactive forms. Classical RSGs, but not atypical RSGs, require regulation by guanine nucleotide exchange factors (GEF), GTPase-activating proteins (GAP) and guanine nucleotide dissociation inhibitors (GDI) to achieve this switch. The objective of this review is to summarize the roles of RSGs in metastatic prostate cancer (mPCa) and their interaction with the androgen receptor (AR), which regulates this disease. Methods: We summarize the literature that describes the role of RSGs in mPCa, and their interaction with the AR. Results: Classical RSGs mostly promote metastasis (except RhoB), whereas atypical RSGs, with exceptions, mostly prevent it. Their role, however, is context-dependent—e.g., RhoB is tumor-suppressive in AR-null PCa but oncogenic in AR-positive tumors. The AR modulates RSG expression transcriptionally, but also affects their function through modulation of GEFs, GAPs, and GDIs. In turn, RSGs also regulate AR transcriptional activity. Interestingly, RSGs and the AR have non-genomic interactions via membrane-localized AR (mAR) not affected by AR inhibitors. Conclusions: Drugs that target RSGs are needed along with AR inhibitors to prevent mPCa progression. Full article
(This article belongs to the Special Issue Advancements in Molecular Research of Prostate Cancer)
Show Figures

Figure 1

17 pages, 1274 KB  
Article
Gene Expression Patterns in Lung Adenocarcinoma Cells in Response to Changes in Deuterium Concentration
by Gabor I. Csonka, András Papp, Ildikó Somlyai and Gábor Somlyai
Int. J. Mol. Sci. 2025, 26(22), 10969; https://doi.org/10.3390/ijms262210969 - 12 Nov 2025
Viewed by 767
Abstract
Deuterium, a stable isotope of hydrogen present in natural water at ~150 ppm, has been implicated in modulating cellular metabolism and tumor progression. While deuterium-depleted water (DDW) has shown anti-cancer effects in preclinical and clinical studies, the underlying transcriptional mechanisms remain incompletely defined. [...] Read more.
Deuterium, a stable isotope of hydrogen present in natural water at ~150 ppm, has been implicated in modulating cellular metabolism and tumor progression. While deuterium-depleted water (DDW) has shown anti-cancer effects in preclinical and clinical studies, the underlying transcriptional mechanisms remain incompletely defined. Here, we profiled gene expression in A549 lung adenocarcinoma cells cultured for 72 h in media containing four graded deuterium concentrations (40, 80, 150, and 300 ppm) using a targeted NanoString panel of 236 cancer-related genes. After stringent quality filtering, 87 genes were retained and classified into nine distinct expression patterns based on fold-change trends relative to the 150 ppm control. High deuterium (300 ppm) induced strong upregulation (up to 2.1-fold) of oncogenic and survival-related genes (e.g., EGFR, CTNNB1, STAT3, CD44), while DDW (40–80 ppm) led to selective downregulation (down to 0.58-fold) of oncogenes (e.g., MYCN, ETS2, IRF1) and drug-resistance genes (e.g., ABCB1). Se-veral genes involved in DNA repair, apoptosis, and extracellular matrix remodeling exhibited dose-dependent responses, suggesting coordinated regulation by deuterium abundance. These findings demonstrate that deuterium concentration functions as a biologically active variable capable of modulating cancer-relevant gene networks. This exploratory dataset refines mechanistic models of DDW action and provides a foundation for future studies incorporating biological replication, functional assays, and in vivo validation. Significance: Deuterium concentration modulation alters oncogenic, apoptotic, and drug-resistance gene networks in lung adenocarcinoma cells, refining prior models of deuterium-depleted water effects. These findings identify deuterium concentration as a biologically active variable warranting further mechanistic and translational investigation. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

16 pages, 1800 KB  
Article
LncRNA-Mediated miR-145 Sponging Drives FN1 and CCND1 Expression: Prognostic and Therapeutic Targets in NSCLC
by Safa Tahmasebi, Davar Amani, Babak Salimi, Ian M. Adcock and Esmaeil Mortaz
Biomolecules 2025, 15(11), 1564; https://doi.org/10.3390/biom15111564 - 6 Nov 2025
Cited by 2 | Viewed by 685
Abstract
Background: Non-small cell lung cancer (NSCLC) progression is driven by dysregulated competing endogenous RNA (ceRNA) networks, where non-coding RNAs sequester miRNAs to modulate oncogene expression. The tumor-suppressor miR-145 is frequently downregulated in NSCLC, but its lncRNA-mediated regulation remains incompletely characterized. Methods: Integrated transcriptomic [...] Read more.
Background: Non-small cell lung cancer (NSCLC) progression is driven by dysregulated competing endogenous RNA (ceRNA) networks, where non-coding RNAs sequester miRNAs to modulate oncogene expression. The tumor-suppressor miR-145 is frequently downregulated in NSCLC, but its lncRNA-mediated regulation remains incompletely characterized. Methods: Integrated transcriptomic analysis of NSCLC datasets (GSE135304: blood RNA from 712 patients; GSE203510: plasma miRNAs) was used to identify dysregulated genes (|log2FC| > 0.1, p < 0.05) and miRNAs (|log2FC| > 1, p < 0.05). Experimentally validated targets from miRTarBase/TarBase were intersected with dysregulated genes, followed by WikiPathways/GO enrichment. ceRNA networks were constructed via co-expression analysis. RT-qPCR validated miR-145-3p expression in A549/MRC-5 cells and NSCLC tissues. GEPIA assessed FN1/CCND1 clinical relevance. Results: We identified 8271 dysregulated genes and 52 miRNAs. miR-145-3p, critical in immune regulation, was significantly downregulated (log2FC = −1.24, p = 0.036). Intersection analysis revealed 27 miR-145-3p targets (e.g., FN1, CCND1, SMAD3) enriched in immune pathways (FDR < 0.05) and TGF-β-mediated EMT within the dysregulated geneset. Six immune-linked hub genes emerged. LncRNAs LOC729919 and LOC100134412 showed strong co-expression with hub genes and competitively bind miR-145-3p, derepressing the expression of the metastasis drivers FN1 (ECM regulator) and CCND1 (cell cycle controller). This ceRNA axis operates within a broader dysregulation of ATM-dependent DNA damage, Hippo signaling, and cell cycle pathways. RT-qPCR confirmed significant miR-145-3p suppression in NSCLC models (p < 0.05). GEPIA revealed a significant FN1-CCND1 co-expression (p = 0.0017). Conclusions: We characterize a novel LOC729919/LOC100134412–miR-145–FN1/CCND1 ceRNA axis in NSCLC pathogenesis. FN1’s prognostic value and functional linkage to CCND1 underscores its potential clinical relevance for therapeutic disruption. Full article
Show Figures

Graphical abstract

27 pages, 2379 KB  
Review
Advances in Understanding Renin–Angiotensin System-Mediated Anti-Tumor Activity of Natural Polyphenols
by Ximing Wu, Mingchuan Yang, Hailing Zhang, Lumin Yang, Yufeng He, Xiaozhong Cheng and Guilan Zhu
Biomolecules 2025, 15(11), 1541; https://doi.org/10.3390/biom15111541 - 2 Nov 2025
Viewed by 762
Abstract
The imbalance of the renin–angiotensin system (RAS), characterized by the overactivation of the pro-tumor ACE/AngII/AT1R axis, is closely linked to tumor growth, angiogenesis, metastasis, and poor prognosis. Natural polyphenols, such as EGCG and resveratrol, exert anti-cancer effects by dual-regulating RAS: they inhibit the [...] Read more.
The imbalance of the renin–angiotensin system (RAS), characterized by the overactivation of the pro-tumor ACE/AngII/AT1R axis, is closely linked to tumor growth, angiogenesis, metastasis, and poor prognosis. Natural polyphenols, such as EGCG and resveratrol, exert anti-cancer effects by dual-regulating RAS: they inhibit the pro-tumor axis by blocking renin, ACE activity, and AT1R expression, while simultaneously activating the protective ACE2/Ang(1-7)/MasR axis. Furthermore, polyphenols and their autoxidation products (e.g., EAOP) modify thiol-containing transmembrane proteins (such as ADAM17 and integrins) and interact with RAS components, further disrupting oncogenic pathways (including MAPK and PI3K/Akt/mTOR) to induce apoptosis, suppress invasion, and reduce oxidative stress. Notably, EAOP exhibits stronger RAS-modulating efficacy than its parent polyphenols. However, challenges such as low bioavailability, insufficient targeting, and limited clinical evidence impede their application. This review provides a comprehensive overview of the anti-cancer mechanisms of polyphenols through RAS regulation, discusses the associated challenges, and proposes potential solutions (including nanodelivery and structural modification) and strategies to advance natural product-based adjuvant treatments. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

13 pages, 1036 KB  
Article
FLI1 Expression in Invasive Breast Carcinoma: Clinicopathological Correlations and Prognostic Implications
by Nusrat Jahan Doly, Dong Yeul Lee, Kazi Nafisa Tahsin, Jhuma Akhter, Shahana Sultana, Julekha Khatun, Sue-zann Chua, A. Tasleema Banu, Qingfeng Chen and Jabed Iqbal
Genes 2025, 16(11), 1313; https://doi.org/10.3390/genes16111313 - 2 Nov 2025
Viewed by 474
Abstract
Background: The E26 transformation-specific (ETS) transcription factor Friend Leukemia Integration 1 (FLI1) has been linked to breast cancer aggressiveness, stromal remodeling, and immune modulation, yet the regulatory mechanisms governing its activity remain poorly defined. Of note, various studies have shown that EWS-FLI1-mediated transcription [...] Read more.
Background: The E26 transformation-specific (ETS) transcription factor Friend Leukemia Integration 1 (FLI1) has been linked to breast cancer aggressiveness, stromal remodeling, and immune modulation, yet the regulatory mechanisms governing its activity remain poorly defined. Of note, various studies have shown that EWS-FLI1-mediated transcription programs are facilitated via direct recruitment and binding of the NuRD-LSD1 complex, regulating its associated gene targets. Furthermore, LSD1 inhibition exhibited reverse transcriptional profiles driven by ETS-FLI and reduced in vivo tumorigenesis in cancers. Methods: We evaluated FLI1 expression across multiple invasive breast carcinoma (IBC) cohorts to determine its prognostic significance and associations with stromal features. In parallel, we investigated FLI1 regulation in humanized breast cancer mouse models treated with an LSD1 inhibitor. Results: High FLI1 expression was associated with advanced histological grade in IBC, consistent with an oncogenic function. FLI1-high tumors also exhibited elevated stromal and immune scores, indicating a role in remodeling the tumor microenvironment. Additionally, LSD1 inhibition downregulated FLI1 target genes involving angiogenesis and invasion. Conclusions: These findings highlight the dual role of FLI1: tumor-intrinsic FLI1 promotes proliferation and invasion, whereas its transcriptional regulation in tumor and endothelial compartments likely reflects LSD1 dependence. Collectively, our results support a mechanistic model in which LSD1–FLI1 crosstalk is involved in immune and stromal remodeling, positioning FLI1 as both a marker of tumor aggressiveness and a potential predictor of response to epigenetic therapies in breast cancer. Full article
(This article belongs to the Special Issue Genetics and Genomics of Human Breast Cancer)
Show Figures

Figure 1

21 pages, 1903 KB  
Article
Evaluation of Expression and Clinicopathological Relevance of Small Nucleolar RNAs (snoRNAs) in Invasive Breast Cancer
by Luděk Záveský, Eva Jandáková, Vít Weinberger, Luboš Minář, Radovan Turyna, Adéla Tefr Faridová, Veronika Hanzíková and Ondřej Slanař
Non-Coding RNA 2025, 11(6), 76; https://doi.org/10.3390/ncrna11060076 - 31 Oct 2025
Viewed by 670
Abstract
Background/Objectives: Breast cancer is a leading cause of cancer-related mortality among women worldwide. Small nucleolar RNAs (snoRNAs) represent a class of non-coding RNAs with potential as novel biomarkers applicable to improve diagnostic and prognostic applications. Methods: We performed a comprehensive evaluation of the [...] Read more.
Background/Objectives: Breast cancer is a leading cause of cancer-related mortality among women worldwide. Small nucleolar RNAs (snoRNAs) represent a class of non-coding RNAs with potential as novel biomarkers applicable to improve diagnostic and prognostic applications. Methods: We performed a comprehensive evaluation of the snoRNA-related gene expression by qPCR using benign and tumor tissue samples associated with invasive breast carcinomas of no special type (NST). Selected candidate snoRNAs, i.e., SCARNA2, SCARNA3, SNORD15B, SNORD94, SNORA68, and SNHG1, along with RNU2-1 snRNA, were further validated and their associations with clinicopathological parameters were examined. External datasets and plasma samples were used for additional validation. Results: SCARNA2 was identified as the most promising snoRNA biomarker candidate, showing a positive association with better progression-free survival (PFS) in our data (13.3-month survival difference between low- and high-expression groups) and with both PFS and overall survival in external RNA-seq datasets. SNORD94, SNORD15B, SCARNA3, and RNU2-1 snRNA were also indicated as putative tumor suppressors. SNORD94 was associated with better progression-free survival (PFS) in our data as well (12.4-month survival difference between low- and high expression groups). Greater downregulation in the low-expression tumor subgroup compared to benign samples further supports the prognostic potential of SCARNA2 and SNORD94. Evidence for SNHG1 and SNORA68 as putative oncogenes was less conclusive. Conclusions: Several small nucleolar RNAs were found to be dysregulated in breast cancer specimens, supporting their further evaluation as potential biomarkers. In particular, SCARNA2, SNORD94, SNORD15B, SCARNA3, and RNU2-1 snRNA merit further investigation to determine their clinical relevance and biological roles in breast cancer. Full article
(This article belongs to the Special Issue Non-coding RNA as Biomarker in Cancer)
Show Figures

Figure 1

11 pages, 269 KB  
Review
A Brief Review on the Role of the Transcription Factor PBX1 in Hematologic Malignancies
by Sofia Chatzileontiadou, Kassiani Boulogeorgou, Christina Frouzaki, Maria Papaioannou, Triantafyllia Koletsa and Evdoxia Hatjiharissi
Int. J. Mol. Sci. 2025, 26(21), 10545; https://doi.org/10.3390/ijms262110545 - 30 Oct 2025
Viewed by 435
Abstract
Pre-B-cell leukemia factor 1 (PBX1) is a transcription factor that plays a significant role in various physiological, developmental, and oncogenic processes in humans. The mechanisms and interactions of PBX1 in both solid and hematologic malignancies remain significant areas of study. It was initially [...] Read more.
Pre-B-cell leukemia factor 1 (PBX1) is a transcription factor that plays a significant role in various physiological, developmental, and oncogenic processes in humans. The mechanisms and interactions of PBX1 in both solid and hematologic malignancies remain significant areas of study. It was initially found in pre-B-cell acute lymphoblastic leukemia as a result of the chromosomal translocation t(1;19). Over the years, its role in other blood neoplasms has been studied. PBX1 and its variant E2A::PBX1 regulate gene expression that influences cell proliferation and differentiation in hematopoietic lineages. Their interaction with oncogenic partners results in abnormal gene regulation and tumorigenesis. Research has predominantly focused on the role of these factors in leukemias and plasma cell neoplasms, whereas other hematologic neoplasms have been largely overlooked. The potential application of PBX1 as a prognostic and predictive biomarker has recently gained attention. However, further research is needed to fully understand its complex role and how it can be targeted for therapeutic purposes. This review summarizes current knowledge on PBX1’s role in the growth of both mature and immature hematologic neoplasms. Moreover, it focuses on its prospective use as a therapeutic target and to predict prognosis, especially for aggressive neoplasms that do not respond to current therapeutic approaches. Full article
(This article belongs to the Section Molecular Biology)
20 pages, 3922 KB  
Article
Identification and Characterization of SRSF2 as a Splicing-Relevant Factor Associated with the Distribution of Membranous to Secreted PD-L1, Exemplarily Considered on Human Renal Tissue, Including Renal Cell Carcinoma
by Tim Hohmann, Urszula Hohmann, Faramarz Dehghani, Hendrik Borgmann, Olaf Grisk, Galyna Pryymachuk and Simon Jasinski-Bergner
Cancers 2025, 17(21), 3463; https://doi.org/10.3390/cancers17213463 - 28 Oct 2025
Viewed by 501
Abstract
Background: The immunomodulatory molecule PD-L1 and its immunological tolerance-mediating interaction with the PD-1 receptor on many immune effector cells represent one of the most important tumor immune checkpoint axes in antibody-based anti-tumor therapies. Furthermore, PD-L1 is subject to alternative splicing, whereby, in addition [...] Read more.
Background: The immunomodulatory molecule PD-L1 and its immunological tolerance-mediating interaction with the PD-1 receptor on many immune effector cells represent one of the most important tumor immune checkpoint axes in antibody-based anti-tumor therapies. Furthermore, PD-L1 is subject to alternative splicing, whereby, in addition to the membrane-bound PD-L1, secreted PD-L1 is also formed as an additional splice variant. This also exerts its effects in the tumor microenvironment, even away from the actual tumor cells, and contributes additional benefits to immune evasion of the tumor. Methods: To examine the association of the splicing factor SRSF2 with the PD-L1 splicing pattern, respective SRSF2 overexpression and knockdown experiments were performed. The precise characterization of SRSF2 followed in human kidney tissue samples and RCCs, including immunofluorescence staining. The impact of the known oncogenic SRSF2 on the host cell transcriptome was further analyzed by RNA sequencing analyses in SRSF2 overexpression and knockdown experiments. Results: In this original research article, the trans splicing factor SRSF2 is identified to be associated with the shift in the alternative splicing pattern of PD-L1 towards the secreted splice variant. The impact of SRSF2 on the cellular transcriptome was demonstrated, and its involvement in the process of malignant transformation, which is obviously also directly linked to immune evasion. Discussion: The optimization of anti-tumor therapies based on monoclonal antibodies against immunomodulatory axes such as PD-1 and PD-L1, including necessary cost reductions, requires the detailed characterization of the gene expression and gene regulation of such molecules, as well as comprehensive molecular biological diagnostics of the tumor sample before putative therapy formulations, e.g., antibody panel collection. Conclusion: Thus, both the amount of PD-L1 protein produced and its splicing pattern are crucial for therapy success and for selecting the most effective therapeutic antibodies. Full article
Show Figures

Figure 1

Back to TopTop