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 (516)

Search Parameters:
Keywords = cell cycle checkpoint

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
30 pages, 13956 KB  
Article
Time- and Dose-Dependent Effects of Irradiation on Endothelial and Tumor Endothelial Cells: Transcriptional, Molecular, and Functional Changes Driving Activation In Vitro and In Vivo
by Iva Santek, Gregor Sersa and Bostjan Markelc
Cancers 2025, 17(17), 2842; https://doi.org/10.3390/cancers17172842 - 29 Aug 2025
Abstract
Background: Irradiation (IR) targets cancer cells, but also the tumor microenvironment, including the tumor’s blood vessels. In addition to tumor endothelial cell (TEC) apoptosis, IR can lead to TEC activation, potentially increasing immune cell infiltration. However, the changes underlying the IR-induced activation of [...] Read more.
Background: Irradiation (IR) targets cancer cells, but also the tumor microenvironment, including the tumor’s blood vessels. In addition to tumor endothelial cell (TEC) apoptosis, IR can lead to TEC activation, potentially increasing immune cell infiltration. However, the changes underlying the IR-induced activation of endothelial cells (ECs) are poorly understood. This study investigated dose- and time-dependent molecular and functional responses of murine and human EC lines to IR in vitro and TECs in vivo in murine tumor models of colorectal carcinoma. Methods: HUVEC, EA.hy926, and Hulec5a, as well as murine bEND.3, 2H11, and SVEC4-10 EC lines, were irradiated with single doses of 2–10 Gy. EC proliferation and survival after IR were assessed by staining all nuclei (Hoechst 33342) and dead cells (propidium iodide) every 24 h for 5 days using the Cytation 1 Cell Imaging Multi-Mode Reader. RNA sequencing analysis of HUVECs irradiated with 2 Gy and 5 Gy at 24 h and 72 h after IR was conducted, focusing on processes related to EC activation. To validate the RNA sequencing results, immunofluorescence staining for proteins related to EC activation, including Stimulator of Interferon Response cGAMP Interactor 1 (STING), Nuclear factor kappa B (NF-κβ), and Vascular cell adhesion molecule 1 (VCAM-1), was performed. To validate the in vitro results, the response of TEC in vivo was analyzed using publicly available RNA sequencing data of TECs isolated from MC38 colon carcinoma irradiated with a single dose of 15 Gy. Finally, murine CT26 colon carcinoma tumors were immunofluorescently stained for STING and NF-κβ 24 and 48 h after IR with a clinically relevant fractionated regimen of 5 × 5 Gy. Results: Doses of 2, 4, 6, 8, and 10 Gy led to a dose-dependent decrease in proliferation and increased death of ECs. RNA sequencing analysis showed that the effects on the transcriptome of HUVECs were most pronounced 72 h after IR with 5 Gy, with 1014 genes (661 down-regulated and 353 up-regulated) being significantly differentially expressed. Irradiation with 5 Gy resulted in HUVEC activation, with up-regulation of the immune system and extracellular matrix genes, such as STING1 (log2FC = 0.81) and SELE (log2FC = 1.09), respectively; and down-regulation of cell cycle markers. Furthermore, IR led to the up-regulation of immune response- and extracellular matrix (ECM)-associated signaling pathways, including NF-κβ signaling and ECM–receptor interaction, which was also observed in the transcriptome of irradiated murine TECs in vivo. This was confirmed at the protein level with higher expressions of the EC activation-associated proteins STING, NF-κβ, and VCAM-1 in irradiated HUVECs and irradiated TECs in vivo. Conclusions: IR induces changes in ECs and TECs, supporting their activation in dose- and time-dependent manners, potentially contributing to the anti-tumor immune response, which may potentially increase the infiltration of immune cells into the tumor and thus, improve the overall efficacy of RT, especially in combination with immune checkpoint inhibitors. Full article
(This article belongs to the Special Issue Radiosensitivity and Radiotoxicity in Cancer)
13 pages, 2086 KB  
Article
Bioactivity-Guided Fractionation and Mechanistic Insights into Aristolochia ringens Root Extract-Induced G1 Phase Arrest and Mitochondria-Mediated Apoptosis in Human Colon Adenocarcinoma Cells
by Saheed O. Anifowose, Abdalrhaman M. Salih, Musa K. Oladejo, Ahmad Rady, Mobarak S. Al Mosallam, Hasan A. Aljohi, Mansour I. Almansour, Saad Hussin Alkahtani, Ibrahim O. Alanazi and Badr A. Al-Dahmash
Pharmaceuticals 2025, 18(9), 1250; https://doi.org/10.3390/ph18091250 - 23 Aug 2025
Viewed by 364
Abstract
Background/Objectives: Aristolochia ringens, a medicinal plant widely used in traditional medicine, has shown potential therapeutic applications. This study aimed to investigate the anticancer mechanism of action of its crude extract against human colorectal adenocarcinoma cells (Caco-2 and HT-29). Methods: Cell [...] Read more.
Background/Objectives: Aristolochia ringens, a medicinal plant widely used in traditional medicine, has shown potential therapeutic applications. This study aimed to investigate the anticancer mechanism of action of its crude extract against human colorectal adenocarcinoma cells (Caco-2 and HT-29). Methods: Cell viability was assessed using the MTT assay to determine IC50 values. Immunofluorescence microscopy was used to examine nuclear morphology and microtubule integrity. Flow cytometry with PI staining was used for cell cycle analysis and Annexin V-FITC/PI staining for apoptosis detection. Mitochondrial membrane potential was evaluated using JC-1 dye. Bioactivity-guided fractionation was performed via HPLC, and GC–MS was used to profile active constituents. Results: The extract exhibited dose-dependent cytotoxicity with IC50 values below 30 µg/mL in colon adenocarcinoma cell lines. Treated Caco-2 cells showed nuclear shrinkage and disrupted microtubules. PI-based flow cytometry revealed G1 phase arrest, and Annexin V-FITC/PI staining indicated enhanced late apoptosis. JC-1 staining demonstrated mitochondrial depolarization. HPLC fractionation identified fractions 2 and 3 as active, and preliminary GC–MS analysis tentatively annotated the presence of alkaloids, sesquiterpenes/diterpenes, and steroidal compounds. Conclusions: A. ringens exerts anticancer effects through a mitochondria-mediated apoptotic pathway, involving G1 checkpoint arrest and cytoskeletal disruption. These findings provide the first integrated cellular and mechanistic evidence of its anticancer potential in colorectal cancer, supporting its promise as a source of novel therapeutic lead compounds. Full article
Show Figures

Graphical abstract

29 pages, 1351 KB  
Review
Molecular Targets for Pharmacotherapy of Head and Neck Squamous Cell Carcinomas
by Robert Sarna, Robert Kubina, Marlena Paździor-Heiske, Adrianna Halama, Patryk Chudy, Paulina Wala, Kamil Krzykawski and Ilona Nowak
Curr. Issues Mol. Biol. 2025, 47(8), 609; https://doi.org/10.3390/cimb47080609 - 1 Aug 2025
Viewed by 411
Abstract
Head and neck squamous cell carcinomas (HNSCCs) represent a heterogeneous group of tumors with a complex molecular profile. Despite therapeutic advances, patient prognosis remains poor, emphasizing the need for more effective treatment strategies. Traditional chemotherapy, with cisplatin and 5-fluorouracil (5-FU), remains the gold [...] Read more.
Head and neck squamous cell carcinomas (HNSCCs) represent a heterogeneous group of tumors with a complex molecular profile. Despite therapeutic advances, patient prognosis remains poor, emphasizing the need for more effective treatment strategies. Traditional chemotherapy, with cisplatin and 5-fluorouracil (5-FU), remains the gold standard but is limited by toxicity and tumor resistance. Immunotherapy, particularly immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and its ligand (PD-L1), has improved overall survival, especially in patients with high PD-L1 expression. In parallel, targeted therapies such as poly (ADP-ribose) polymerase 1 (PARP1) inhibitors—which impair DNA repair and increase replication stress—have shown promising activity in HNSCC. Cyclin-dependent kinase (CDK) inhibitors are also under investigation due to their potential to correct dysregulated cell cycle control, a hallmark of HNSCC. This review aims to summarize current and emerging pharmacotherapies for HNSCC, focusing on chemotherapy, immunotherapy, and PARP and CDK inhibitors. It also discusses the evolving role of targeted therapies in improving clinical outcomes. Future research directions include combination therapies, nanotechnology-based delivery systems to enhance treatment specificity, and the development of diagnostic tools such as PARP1-targeted imaging to better guide personalized treatment approaches. Full article
(This article belongs to the Special Issue Future Challenges of Targeted Therapy of Cancers: 2nd Edition)
Show Figures

Figure 1

36 pages, 5612 KB  
Review
The Multifaceted Role of p53 in Cancer Molecular Biology: Insights for Precision Diagnosis and Therapeutic Breakthroughs
by Bolong Xu, Ayitila Maimaitijiang, Dawuti Nuerbiyamu, Zhengding Su and Wenfang Li
Biomolecules 2025, 15(8), 1088; https://doi.org/10.3390/biom15081088 - 27 Jul 2025
Viewed by 1017
Abstract
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and [...] Read more.
The protein p53, often referred to as the “guardian of the genome,” is essential for preserving cellular balance and preventing cancerous transformations. As one of the most commonly altered genes in human cancers, its impaired function is associated with tumor initiation, development, and resistance to treatment. Exploring the diverse roles of p53, which include regulating the cell cycle, repairing DNA, inducing apoptosis, reprogramming metabolism, and modulating immunity, provides valuable insights into cancer mechanisms and potential treatments. This review integrates recent findings on p53′s dual nature, functioning as both a tumor suppressor and an oncogenic promoter, depending on the context. Wild-type p53 suppresses tumors by inducing cell cycle arrest or apoptosis in response to genotoxic stress, while mutated variants often lose these functions or gain novel pro-oncogenic activities. Emerging evidence highlights p53′s involvement in non-canonical pathways, such as regulating tumor microenvironment interactions, metabolic flexibility, and immune evasion mechanisms. For instance, p53 modulates immune checkpoint expression and influences the efficacy of immunotherapies, including PD-1/PD-L1 blockade. Furthermore, advancements in precision diagnostics, such as liquid biopsy-based detection of p53 mutations and AI-driven bioinformatics tools, enable early cancer identification and stratification of patients likely to benefit from targeted therapies. Therapeutic strategies targeting p53 pathways are rapidly evolving. Small molecules restoring wild-type p53 activity or disrupting mutant p53 interactions, such as APR-246 and MDM2 inhibitors, show promise in clinical trials. Combination approaches integrating gene editing with synthetic lethal strategies aim to exploit p53-dependent vulnerabilities. Additionally, leveraging p53′s immunomodulatory effects through vaccine development or adjuvants may enhance immunotherapy responses. In conclusion, deciphering p53′s complex biology underscores its unparalleled potential as a biomarker and therapeutic target. Integrating multi-omics analyses, functional genomic screens, and real-world clinical data will accelerate the translation of p53-focused research into precision oncology breakthroughs, ultimately improving patient outcomes. Full article
(This article belongs to the Special Issue DNA Damage and Repair in Cancer Treatment)
Show Figures

Figure 1

18 pages, 2432 KB  
Article
High Carbon Dioxide Concentration Inhibits Pileus Growth of Flammulina velutipes by Downregulating Cyclin Gene Expression
by Kwan-Woo Lee, Che-Hwon Park, Seong-Chul Lee, Ju-Hyeon Shin and Young-Jin Park
J. Fungi 2025, 11(8), 551; https://doi.org/10.3390/jof11080551 - 24 Jul 2025
Viewed by 480
Abstract
Flammulina velutipes is a widely cultivated edible mushroom in East Asia, recognized for its nutritional benefits and distinct morphology characterized by a long stipe and a compact, hemispherical pileus. The pileus not only plays a critical biological role in reproduction through spore formation [...] Read more.
Flammulina velutipes is a widely cultivated edible mushroom in East Asia, recognized for its nutritional benefits and distinct morphology characterized by a long stipe and a compact, hemispherical pileus. The pileus not only plays a critical biological role in reproduction through spore formation but also serves as a key commercial trait influencing consumer preference and market value. Despite its economic importance, pileus development in F. velutipes is highly sensitive to environmental factors, among which carbon dioxide (CO2) concentration is particularly influential under indoor cultivation conditions. While previous studies have reported that elevated CO2 levels can inhibit pileus expansion in other mushroom species, the molecular mechanisms by which CO2 affects pileus growth in F. velutipes remain poorly understood. In this study, we investigated the impact of CO2 concentration on pileus morphology and gene expression in F. velutipes by cultivating fruiting bodies under two controlled atmospheric conditions: low (1000 ppm) and high (10,000 ppm) CO2. Morphometric analysis revealed that elevated CO2 levels significantly suppressed pileus expansion, reducing the average diameter by more than 50% compared to the low CO2 condition. To elucidate the underlying genetic response, we conducted RNA sequencing and identified 102 differentially expressed genes (DEGs), with 78 being downregulated under elevated CO2. Functional enrichment analysis highlighted the involvement of cyclin-dependent protein kinase regulatory pathways in this response. Two cyclin genes were found to be significantly downregulated under elevated CO2 conditions, and their suppression was validated through quantitative real-time PCR. These genes, possessing conserved cyclin_N domains, are implicated in the regulation of the eukaryotic cell cycle, particularly in mitotic growth. These results indicate that CO2-induced downregulation of cyclin genes may underlie cell cycle arrest, contributing to inhibited pileus development. This study is the first to provide transcriptomic evidence that elevated CO2 concentrations specifically repress PHO80-like cyclin genes in F. velutipes, revealing a molecular mechanism by which CO2 stress inhibits pileus development. These findings suggest that elevated CO2 triggers a morphogenetic checkpoint by repressing PHO80-like cyclins, thereby modulating cell cycle progression during fruiting body development. This study provides the first evidence of such a transcriptional response in edible mushrooms and offers promising molecular targets for breeding CO2-resilient strains and optimizing commercial cultivation conditions. Full article
(This article belongs to the Special Issue Molecular Biology of Mushroom)
Show Figures

Figure 1

13 pages, 1431 KB  
Communication
Glucocorticoids Downregulate PD-L1 in Glioblastoma Cells via GILZ-Mediated ERK Inhibition
by Sabrina Adorisio, Giorgia Renga, Domenico Vittorio Delfino and Emira Ayroldi
Biomedicines 2025, 13(8), 1793; https://doi.org/10.3390/biomedicines13081793 - 22 Jul 2025
Viewed by 315
Abstract
Glucocorticoids (GCs), such as dexamethasone (DEX), are commonly administered to glioblastoma (GBM) patients to control cerebral edema; however, their effects on immune checkpoint regulation in tumor cells remain insufficiently characterized. This study examined the impact of DEX on the expression of programmed death-ligand [...] Read more.
Glucocorticoids (GCs), such as dexamethasone (DEX), are commonly administered to glioblastoma (GBM) patients to control cerebral edema; however, their effects on immune checkpoint regulation in tumor cells remain insufficiently characterized. This study examined the impact of DEX on the expression of programmed death-ligand 1 (PD-L1) and glucocorticoid-induced leucine zipper (GILZ), a downstream effector of glucocorticoid receptor (GR) signaling, in the U87 and U251 glioblastoma cell lines. DEX consistently induced GILZ expression in both models yet elicited divergent effects on PD-L1: suppression in U87 cells and upregulation in U251 cells. In U87 cells, DEX-induced PD-L1 downregulation was accompanied by accelerated cell cycle progression, suggesting a dual impact on tumor immune evasion and proliferation. Mechanistically, GILZ silencing restored ERK phosphorylation and reversed PD-L1 suppression, whereas GILZ overexpression further decreased PD-L1 levels, implicating a GILZ–ERK pathway in the control of PD-L1. These findings uncover a previously unrecognized GR–GILZ–PD-L1 regulatory axis in glioblastoma cells. While these results are based on in vitro models, they provide a rationale for future in vivo studies to determine whether modulation of GILZ may influence immune checkpoint dynamics and therapeutic responsiveness in GBM. Full article
Show Figures

Figure 1

21 pages, 5260 KB  
Review
Disrupting Cell Cycle Machinery: CREPT Is an Emerging Target in Cancer Therapy
by Umar Farooq, Jun Li and Zhijie Chang
Cancers 2025, 17(14), 2401; https://doi.org/10.3390/cancers17142401 - 19 Jul 2025
Viewed by 853
Abstract
The transcriptional co-factor cell-cycle-related and expression-elevated protein in tumors (CREPT) has emerged as a critical driver of the cell cycle and a significant contributor to tumorigenesis. The aberrant expression or upregulation of CREPT boosts multiple signaling pathways, including Wnt/β-catenin, STAT3 and NF-κB/TNFR2, which [...] Read more.
The transcriptional co-factor cell-cycle-related and expression-elevated protein in tumors (CREPT) has emerged as a critical driver of the cell cycle and a significant contributor to tumorigenesis. The aberrant expression or upregulation of CREPT boosts multiple signaling pathways, including Wnt/β-catenin, STAT3 and NF-κB/TNFR2, which are frequently dysregulated in various cancers and are associated with poor overall survival. In preclinical studies, CREPT knockdown via shRNA has demonstrated sustained tumor growth regression. Recent researches have uncovered additional functions of CREPT, including roles in metabolic regulation, tissue repair, and microenvironmental remodeling, further establishing it as a pleiotropic transcriptional regulator. Currently, there is no therapeutic agent that directly inhibits CREPT expression in clinic. However, miRNAs and other methods have been used to target CREPT, which have yielded useful results in inhibiting tumor growth. In this review, we discuss the role of CREPT in the hallmarks of cancer and propose that targeting CREPT will reverse tumor growth and may improve the immune checkpoint inhibitors in combination in CREPT-driven cancers. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Graphical abstract

14 pages, 1865 KB  
Article
Plasma WFDC2 (HE4) as a Predictive Biomarker for Clinical Outcomes in Cancer Patients Receiving Anti-PD-1 Therapy: A Pilot Study
by Makoto Watanabe, Katsuaki Ieguchi, Takashi Shimizu, Ryotaro Ohkuma, Risako Suzuki, Emiko Mura, Nana Iriguchi, Tomoyuki Ishiguro, Yuya Hirasawa, Go Ikeda, Masahiro Shimokawa, Hirotsugu Ariizumi, Kiyoshi Yoshimura, Atsushi Horiike, Takuya Tsunoda, Mayumi Tsuji, Shinichi Kobayashi, Tatsunori Oguchi, Yuji Kiuchi and Satoshi Wada
Cancers 2025, 17(14), 2384; https://doi.org/10.3390/cancers17142384 - 18 Jul 2025
Viewed by 484
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy; however, reliable biomarkers of therapeutic efficacy remain limited. We investigated the clinical utility of plasma WFDC2 levels in patients receiving anti-PD-1 antibody treatment. Methods: Twenty-one patients with non-small cell lung, gastric, or [...] Read more.
Background/Objectives: Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy; however, reliable biomarkers of therapeutic efficacy remain limited. We investigated the clinical utility of plasma WFDC2 levels in patients receiving anti-PD-1 antibody treatment. Methods: Twenty-one patients with non-small cell lung, gastric, or bladder cancer received nivolumab or pembrolizumab. Plasma WFDC2 concentrations were measured by ELISA before ICI treatment (pre-ICI) and after two and four treatment cycles. Associations between WFDC2 expression changes and overall survival (OS), progression-free survival (PFS), and tumor progression were assessed. ROC curve analyses compared the predictive performance of WFDC2, soluble PD-L1 (sPD-L1), soluble PD-1 (sPD-1), and their combinations, with the area under the curve (AUC) evaluating predictive accuracy. Results: Levels of WFDC2 pre-ICI and those after two cycles were significantly higher than levels in healthy donors. However, no significant differences in WFDC2 levels were found between the time points during treatment. Greater increases in WFDC2 levels were significantly correlated with shorter OS (p = 0.002), shorter PFS (p = 0.037), and tumor progression (p = 0.003). ROC analysis revealed that WFDC2 achieved a higher AUC (0.700) than sPD-L1 (0.538) or sPD-1 (0.650). Combining biomarkers improved the predictive accuracy, with sPD-L1 plus WFDC2 showing the highest AUC (0.825). Conclusions: Serial increases in plasma WFDC2 are associated with poor clinical outcomes, highlighting its potential as a biomarker. Baseline plasma WFDC2 outperformed sPD-L1 and sPD-1 diagnostically. These findings should be interpreted as exploratory and hypothesis-generating, requiring confirmation in larger, tumor-specific cohorts with multivariate adjustment. WFDC2 represents a promising minimally invasive biomarker for the early identification of patients unlikely to benefit from ICI therapy. Full article
Show Figures

Figure 1

27 pages, 7413 KB  
Article
The Effect of the Ethanolic Extracts from Syzygium aromaticum and Syzygium nervosum on Antiproliferative Activity and Apoptosis in HCT116 and HT-29 Cells
by Thunyatorn Yimsoo, Weerakit Taychaworaditsakul, Sunee Chansakaow, Sumet Kongkiatpaiboon, Ngampuk Tayana, Teera Chewonarin, Parirat Khonsung and Seewaboon Sireeratawong
Int. J. Mol. Sci. 2025, 26(14), 6826; https://doi.org/10.3390/ijms26146826 - 16 Jul 2025
Viewed by 425
Abstract
Colorectal cancer (CRC) is the third most diagnosed cancer worldwide, and p53 dysfunction plays a significant role in its pathogenesis by impairing cell cycle control and apoptosis. This study aimed to elucidate the phytochemical composition and anticancer potential of extract of residue from [...] Read more.
Colorectal cancer (CRC) is the third most diagnosed cancer worldwide, and p53 dysfunction plays a significant role in its pathogenesis by impairing cell cycle control and apoptosis. This study aimed to elucidate the phytochemical composition and anticancer potential of extract of residue from clove hydrodistillation (Syzygium aromaticum, SA) and seed extract from Syzygium nervosum (SN). LC-DAD-MS/MS analysis identified gallic acid (2.68%) and ellagic acid (6.70%) as major constituents in SA, while SN contained gallic acid (0.26%), ellagic acid (3.06%), and 2′,4′-dihydroxy-6′-methoxy-3′,5′-dimethylchalcone (DMC) as major constituents. Both extracts exhibited potent antioxidant effects as evidenced by DPPH and ABTS assays. In vitro assays showed that SA and SN significantly inhibited the proliferation of HCT116 (p53 wild-type) colorectal cancer cells, with minimal effects on HT-29 (p53 mutant) cells. Apoptosis was confirmed in HCT116 via Annexin V-FITC/PI staining and increased caspase-3/7 activity. Cell cycle analysis revealed sub-G1 accumulation, accompanied by upregulated p21 and concurrently downregulated cyclin D1 expression, both hallmarks of p53-mediated checkpoint activation. These molecular effects were not observed in HT-29 cells. In conclusion, SA and SN extracts selectively induce apoptosis and cell cycle arrest in p53-functional CRC cells, likely mediated by their phenolic constituents. These findings support their potential as promising plant-derived therapeutic agents for targeted colorectal cancer treatment. Full article
(This article belongs to the Special Issue Molecular Research and Potential Effects of Medicinal Plants)
Show Figures

Figure 1

40 pages, 2429 KB  
Review
Hepatocytes as Model for Investigating Natural Senotherapeutic Compounds and Their Effects on Cell Cycle Dynamics and Genome Stability
by Anastasia Fizikova, Anna Prokhorova, Daria Churikova, Zahar Konstantinov, Roman Ivanov, Alexander Karabelsky and Stanislav Rybtsov
Int. J. Mol. Sci. 2025, 26(14), 6794; https://doi.org/10.3390/ijms26146794 - 16 Jul 2025
Viewed by 1037
Abstract
DNA is inherently unstable and is susceptible to damage from both endogenous sources (such as reactive oxygen species) and exogenous factors (including UV, ionizing radiation, and chemicals). The accumulation of DNA damage manifests as genetic mutations, chromosomal instability, and the stalling of DNA [...] Read more.
DNA is inherently unstable and is susceptible to damage from both endogenous sources (such as reactive oxygen species) and exogenous factors (including UV, ionizing radiation, and chemicals). The accumulation of DNA damage manifests as genetic mutations, chromosomal instability, and the stalling of DNA replication and transcription processes. Accumulated DNA damage influences apoptosis and cell cycle checkpoints, serving as one of the key triggers for the manifestation of the senescent phenotype. Both aging and cancer are associated with the accumulation of mutations in somatic cells. Disruption of cell cycle control and uncontrolled proliferation are fundamental characteristics of any cancer cell, with the majority of anticancer drugs acting as inhibitors of cyclin-dependent kinases, thereby inducing a transition of cells into a senescent state. Consequently, disturbances in the dynamics and regulation of inflammatory responses, oxidative stress, cell proliferation, DNA damage repair, and epigenetic anomalies, along with the influence of retroviruses and transposons, lead to the accumulation of senescent cells within the human body, characterized by blocked replication and cell cycle, as well as a distinct secretory phenotype. The age-related or disease-associated accumulation of these senescent cells significantly alters the physiology of tissues and the organism as a whole. Many secondary metabolites of higher plants exhibit senolytic and senomorphic activities, although most of them are not fully characterized. In this review, we will explore the principal signaling pathways in mammalian cells that govern the cell cycle and cellular senescence, with a particular emphasis on how their dynamics, expression, and regulation have been modified through the application of senotherapeutic compounds. The second section of the review will identify key target genes for the metabolic engineering, primarily aimed at enhancing the accumulation of plant secondary metabolites with potential therapeutic benefits. Lastly, we will discuss the rationale for utilizing liver cells as a model system to investigate the effects of senolytic compounds on human physiology and health, as well as how senotherapeutic substances can be leveraged to improve gene therapy approaches based on CRISPR/Cas9 and prime-editing technologies. Full article
(This article belongs to the Collection State-of-the-Art Macromolecules in Russia)
Show Figures

Figure 1

18 pages, 11393 KB  
Article
Expression Characteristics and Prognostic Value of KLRG2 in Endometrial Cancer: A Comprehensive Analysis Based on Multi-Omics Data
by Xiaoyan Huang, Ailian Li and Dianbo Xu
Biomedicines 2025, 13(7), 1592; https://doi.org/10.3390/biomedicines13071592 - 30 Jun 2025
Viewed by 510
Abstract
Background: Endometrial cancer (EC) remains a major gynecologic malignancy with limited biomarkers for risk stratification. While killer cell lectin-like receptor G2 (KLRG2) exhibits oncogenic properties in other cancers, its clinical significance and mechanistic roles in EC are unknown. This study aims to [...] Read more.
Background: Endometrial cancer (EC) remains a major gynecologic malignancy with limited biomarkers for risk stratification. While killer cell lectin-like receptor G2 (KLRG2) exhibits oncogenic properties in other cancers, its clinical significance and mechanistic roles in EC are unknown. This study aims to systematically characterize KLRG2 expression in EC, evaluate its prognostic significance, decipher underlying molecular mechanisms, and explore its role in tumor immune microenvironment regulation. Methods: We performed integrated multi-omics analyses using TCGA-UCEC (n = 552), GTEx, and GEO cohorts (GSE106191), complemented by qPCR validation (14 EC vs. 14 normal samples). Prognostic models were constructed via Cox regression and time-dependent ROC analysis. Epigenetic regulation was assessed through methylation profiling (UALCAN/MethSurv), and immune correlations were evaluated using TIMER/ESTIMATE algorithms. Results: KLRG2 was significantly overexpressed in EC tissues compared to normal endometrium (p < 0.001), validated by immunohistochemistry and qPCR. High KLRG2 expression independently predicted worse overall survival (HR = 3.08, 95% CI = 1.92–4.96) and progression-free interval (HR = 1.98, 95% CI = 1.37–2.87). Furthermore, elevated KLRG2 levels were significantly associated with advanced-stage disease (p < 0.001), deep myometrial invasion (p < 0.05), and high-grade histology (p < 0.001). Mechanistically, promoter hypomethylation was associated with KLRG2 overexpression (p < 0.001), while hypermethylation at three CpG sites (cg04915254, cg04520485, cg23104233) correlated with poor prognosis. Functional enrichment linked KLRG2 to cell cycle checkpoints and G Protein-Coupled Receptor signaling. Immune profiling revealed cytotoxic lymphocyte depletion (CD8+ T cells: Spearman’s ρ = −0.247, p < 0.001; NK CD56bright cells: Spearman’s ρ = −0.276, p < 0.001) and Th2 polarization (Spearman’s ρ = 0.117, p = 0.006). Conclusions: This comprehensive EC study establishes KLRG2 as a dual diagnostic/prognostic biomarker and immunomodulatory target. These findings provide a rationale for developing KLRG2-directed therapies to counteract tumor-intrinsic proliferation and microenvironmental immune suppression. Future single-cell analyses are warranted to dissect KLRG2-mediated tumor-immune crosstalk. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

11 pages, 606 KB  
Systematic Review
Salvage Pulmonary Resection After Immune Checkpoint or Tyrosine Kinase Inhibitor Therapy for Initially Unresectable Non-Small-Cell Lung Cancer: A Systematic Review
by Vasile Gaborean, Catalin Vladut Ionut Feier, Razvan Constantin Vonica, Alaviana Monique Faur, Vladut Iosif Rus and Calin Muntean
Biomedicines 2025, 13(7), 1541; https://doi.org/10.3390/biomedicines13071541 - 24 Jun 2025
Viewed by 536
Abstract
Background and Objectives: Systemic conversion of stage III–IV non-small-cell lung cancer (NSCLC) to a surgically resectable state with immune checkpoint inhibitors (ICIs) or tyrosine kinase inhibitors (TKIs) creates an emerging cohort of candidates for “salvage” pulmonary resection. No comprehensive evidence synthesis has yet [...] Read more.
Background and Objectives: Systemic conversion of stage III–IV non-small-cell lung cancer (NSCLC) to a surgically resectable state with immune checkpoint inhibitors (ICIs) or tyrosine kinase inhibitors (TKIs) creates an emerging cohort of candidates for “salvage” pulmonary resection. No comprehensive evidence synthesis has yet evaluated the feasibility, safety, or oncologic value of this strategy. We aimed to systematically review peri-operative and survival outcomes of salvage lung resection following ICI or TKI therapy. Methods: MEDLINE, Embase, and PubMed were searched (inception–1 May 2025). Studies reporting ≥5 adult NSCLC patients who underwent anatomical lung resection after at least one cycle of ICI or TKI were eligible. Two reviewers screened records, extracted predefined variables, and assessed risk of bias with the Newcastle–Ottawa Scale. Pooled proportions were calculated with a random-effects model. Results: Fourteen observational series (n = 312 patients) met inclusion. Median age was 62 years (range 38–81); 58% were male. Lobectomy (63%) and segmentectomy (21%) were most frequent. Video-assisted/robotic approaches were achieved in 48%. The pooled R0 rate was 93% (95% CI 89–97%); pathologic complete response occurred in 27% (95% CI 19–36%). Major complications (Clavien–Dindo ≥ III) were 11% (95% CI 7–16%), and 30-day mortality was 1.3% (95% CI 0–3%). One-year disease-free and overall survival were 68% and 88%, respectively. Conclusions: Current evidence—albeit heterogeneous—indicates that salvage pulmonary resection after modern systemic conversion therapy is technically feasible, associated with acceptably low morbidity, and yields encouraging short-term oncologic outcomes. Prospective, registry-based studies are needed to define selection criteria and long-term benefit. Full article
Show Figures

Figure 1

22 pages, 2036 KB  
Review
Radiogenomics of Stereotactic Radiotherapy: Genetic Mechanisms Underlying Radiosensitivity, Resistance, and Immune Response
by Damir Vučinić, Ana-Marija Bukovica Petrc, Ivona Antončić, Maja Kolak Radojčić, Matea Lekić and Felipe Couñago
Genes 2025, 16(7), 732; https://doi.org/10.3390/genes16070732 - 24 Jun 2025
Viewed by 1190
Abstract
Stereotactic body radiotherapy (SBRT) delivers ablative radiation doses with sub-millimeter precision. Radiogenomic studies, meanwhile, provide insights into how tumor-intrinsic genetic factors influence responses to such high-dose treatments. This review explores the radiobiological mechanisms underpinning SBRT efficacy, emphasizing the roles of DNA damage response [...] Read more.
Stereotactic body radiotherapy (SBRT) delivers ablative radiation doses with sub-millimeter precision. Radiogenomic studies, meanwhile, provide insights into how tumor-intrinsic genetic factors influence responses to such high-dose treatments. This review explores the radiobiological mechanisms underpinning SBRT efficacy, emphasizing the roles of DNA damage response (DDR) pathways, tumor suppressor gene alterations, and inflammatory signaling in shaping tumor radiosensitivity or resistance. SBRT induces complex DNA double-strand breaks (DSBs) that robustly activate DDR signaling cascades, particularly via the ATM and ATR kinases. Tumors with proficient DNA repair capabilities often resist SBRT, whereas deficiencies in key repair genes can render them more susceptible to radiation-induced cytotoxicity. Mutations in tumor suppressor genes may impair p53-dependent apoptosis and disrupt cell cycle checkpoints, allowing malignant cells to evade radiation-induced cell death. Furthermore, SBRT provokes the release of pro-inflammatory cytokines and activates innate immune pathways, potentially leading to immunogenic cell death and reshaping the tumor microenvironment. Radiogenomic profiling has identified genomic alterations and molecular signatures associated with differential responses to SBRT and immune activation. These insights open avenues for precision radiotherapy approaches, including the use of genomic biomarkers for patient selection, the integration of SBRT with DDR inhibitors or immunotherapies, and the customization of treatment plans based on individual tumor genotypes and immune landscapes. Ultimately, these strategies aim to enhance SBRT efficacy and improve clinical outcomes through biologically tailored treatment. This review provides a comprehensive summary of current knowledge on the genetic determinants of response to stereotactic radiotherapy and discusses their implications for personalized cancer treatment. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

24 pages, 2487 KB  
Review
Targeting WEE1 Kinase for Breast Cancer Therapeutics: An Update
by Zhao Zhang, Ritika Harish, Naveed Elahi, Sawanjit Saini, Aamir Telia, Manjit Kundlas, Allexes Koroleva, Israel N. Umoh, Manpreet Lota, Meha Bilkhu, Aladdin Kawaiah, Manogna R. Allala, Armelle Leukeu, Emmanuel Nebuwa, Nadiya Sharifi, Anthony W. Ashton, Xuanmao Jiao and Richard G. Pestell
Int. J. Mol. Sci. 2025, 26(12), 5701; https://doi.org/10.3390/ijms26125701 - 13 Jun 2025
Viewed by 2163
Abstract
WEE1 kinase is a crucial cell cycle regulatory protein that controls the timing of mitotic entry. WEE1, via inhibition of Cyclin-dependent Kinase 1 (CDK1) and Cyclin-dependent Kinase 2 (CDK2), governs the G2-M checkpoint by inhibiting entry into mitosis. The state of balance between [...] Read more.
WEE1 kinase is a crucial cell cycle regulatory protein that controls the timing of mitotic entry. WEE1, via inhibition of Cyclin-dependent Kinase 1 (CDK1) and Cyclin-dependent Kinase 2 (CDK2), governs the G2-M checkpoint by inhibiting entry into mitosis. The state of balance between WEE family kinases and CDC25C phosphatases restricts CDK1/CycB activity. The WEE kinase family consists of WEE1, PKMYT1, and WEE2 (WEE1B). WEE1 and PKMYT1 regulate entry into mitosis during cell cycle progression, whereas WEE2 governs cell cycle progression during meiosis. Recent studies have identified WEE1 as a potential therapeutic target in several cancers, including therapy-resistant triple-negative breast cancer. Adavosertib’s clinical promise was challenged by inter-individual variations in response and side effects. Because of these promising preclinical outcomes, other WEE1 kinase inhibitors (Azenosertib, SC0191, IMP7068, PD0407824, PD0166285, WEE1-IN-5, Zedoresertib, WEE1-IN-8, and ATRN-1051) are being developed, with several currently being evaluated in clinical trials or as an adjuvant to chemotherapies. Preclinical studies show WEE1 inhibitors induce MHC class 1 antigens and STING when given as combination therapies, suggesting potential additional therapeutic opportunities. Reliable predictors of clinical responses based on mechanistic insights remain an important unmet need. Herein, we review the role of WEE1 inhibition therapy in breast cancer. Full article
(This article belongs to the Special Issue Molecular Research and Treatment of Breast Cancer: 3rd Edition)
Show Figures

Figure 1

18 pages, 938 KB  
Article
Periodic Boosters of COVID-19 Vaccines Do Not Affect the Safety and Efficacy of Immune Checkpoint Inhibitors for Advanced Non-Small Cell Lung Cancer: A Longitudinal Analysis of the Vax-On-Third Study
by Agnese Fabbri, Enzo Maria Ruggeri, Antonella Virtuoso, Diana Giannarelli, Armando Raso, Fabrizio Chegai, Daniele Remotti, Carlo Signorelli and Fabrizio Nelli
Cancers 2025, 17(12), 1948; https://doi.org/10.3390/cancers17121948 - 11 Jun 2025
Viewed by 1023
Abstract
Background: Increasing evidence suggests that the immunogenicity of COVID-19 mRNA vaccines might influence the efficacy of immune checkpoint inhibitors (ICIs). Current studies have not considered the impact of additional vaccinations, which are now recommended as a preventive strategy against SARS-CoV-2 infection for cancer [...] Read more.
Background: Increasing evidence suggests that the immunogenicity of COVID-19 mRNA vaccines might influence the efficacy of immune checkpoint inhibitors (ICIs). Current studies have not considered the impact of additional vaccinations, which are now recommended as a preventive strategy against SARS-CoV-2 infection for cancer patients receiving active treatments. Consequently, we leveraged the prospective monitoring from the Vax-On-Third study to explore whether periodic mRNA vaccine boosters administered around the start of ICIs could influence the rates of immune-related adverse events (irAEs) and survival outcomes in patients with advanced non-small cell lung cancer (NSCLC). Methods: Our study included patients with a histological diagnosis of metastatic NSCLC and available PD-L1 tumor proportion score (TPS), who had undergone at least two cycles of upfront treatment with pembrolizumab, cemiplimab, or their combination with platinum-based chemotherapy. Patients who received any additional mRNA-based vaccine doses within 60 days before to 30 days after starting ICIs accounted for the exposed cohort. Those who declined further boosters formed the reference cohort. We analyzed differences in irAE frequencies, progression-free survival (PFS), and overall survival (OS) using univariate and multivariate analyses. Results: Between 27 November 2021 and 31 March 2024, we enrolled 226 eligible patients. The exposed cohort consisted of 112 patients who had received either a third or fourth dose of tozinameran or a bivalent booster. Based on PD-L1 expression levels, 93 (41%) and 133 (59%) patients received single-agent ICIs (PD-L1 TPS ≥ 50%) or combination regimens (PD-L1 TPS < 50%), respectively. Propensity-score matching using comprehensive criteria resulted in two cohorts of 102 patients each, with an optimal balance of prognostic factors. A thorough analysis of any grade irAEs showed no significant differences between the cohorts. A longitudinal survival assessment with a median follow-up of 22.8 (95% CI 19.2–26.0) months showed no difference between the cohorts. The median PFS for the reference and exposed cohorts was 7.5 (95% CI 5.9–9.1) and 8.2 (95% CI 6.2–10.2) months, respectively (p = 0.408; HR 0.88 [95% CI 0.66–1.18]). The median OS for the reference and exposed cohorts was 10.5 (95% CI 7.9–13.0) and 13.8 (95% CI 12.0–15.5) months, respectively (p = 0.170; HR 0.81 [95% CI 0.59–1.09]). Multivariate analysis confirmed that receiving additional mRNA vaccine boosters did not significantly affect the risk of disease progression or mortality. Univariate analysis within the subgroup of patients with high PD-L1 TPS who received single-agent ICIs showed a significant OS advantage for patients in the exposed cohort (9.7 [95% CI 8.1–11.2] vs. 18.6 [95% CI 13.5–23.6] months; p = 0.034; HR 0.59 [95% CI 0.36–0.96]). Conclusion: After optimally balancing prognostic factors, regular mRNA vaccine boosters at the onset of ICIs did not impact the safety and survival of patients with advanced NSCLC. The improved outcome observed in patients with high PD-L1 expression levels aligns with previous findings and warrants further investigation. Full article
Show Figures

Figure 1

Back to TopTop