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

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
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,324)

Search Parameters:
Keywords = G2 arrest

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 2086 KiB  
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 (registering DOI) - 23 Aug 2025
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

21 pages, 3094 KiB  
Article
Anticancer Activity of Ethanolic Extract of Tabernaemontana catharinensis in Breast Cancer Lines MCF-7 and MDA-MB-231
by Diana del Carmen Martínez-Méndez, María de la Luz Sánchez-Mundo, María del Rocío Thompson-Bonilla, Luis Marat Álvarez-Salas, Víctor Hugo Rosales-García, Jacobo Rodríguez-Campos and María Eugenia Jaramillo-Flores
Int. J. Mol. Sci. 2025, 26(16), 8111; https://doi.org/10.3390/ijms26168111 - 21 Aug 2025
Abstract
Breast cancer is a serious public health problem worldwide. Although current treatments with drugs such as cisplatin and paclitaxel are effective, they are associated with severe adverse effects and the development of drug resistance, which has prompted the search for new therapeutic strategies. [...] Read more.
Breast cancer is a serious public health problem worldwide. Although current treatments with drugs such as cisplatin and paclitaxel are effective, they are associated with severe adverse effects and the development of drug resistance, which has prompted the search for new therapeutic strategies. In this context, the present study evaluated the anticancer activity of the ethanolic extract of Tabernaemontana catharinensis (EET) on the breast cancer cell lines MCF-7 (hormone-sensitive) and MDA-MB-231 (triple-negative) using 2D and 3D models. The results showed that EET significantly reduced cell viability in both lines, with IC50 values of 83.06 µg/mL (MCF-7) and 8.3 µg/mL (MDA-MB-231) in 2D and 499.3 µg/mL and 280 µg/mL, respectively, in 3D. In addition, treatment with EET caused cell cycle arrest in the G1 phase, reduced CDK4 activity by 58% and ALDH3A1 activity by 32%, and increased levels of the tumor suppressor protein p53. Significant induction of apoptosis was also observed, evidenced by the activation of caspases -3/7, -8, and -9, along with a decrease in intracellular ATP levels (37% in MCF-7 and 90% in MDA-MB-231), suggesting mitochondrial dysfunction. Finally, EET showed the ability to inhibit cell invasion. Taken together, these results indicate that the ethanolic extract of Tabernaemontana catharinensis has potent antiproliferative, proapoptotic, and antimetastatic activity in breast cancer cells, in both two-dimensional and three-dimensional models. Its effect on various key molecular pathways and its ability to enhance the action of conventional chemotherapeutic agents position it as a promising adjuvant agent in the treatment of breast cancer. Full article
(This article belongs to the Special Issue Natural Products in Cancer Prevention and Treatment)
Show Figures

Graphical abstract

12 pages, 355 KiB  
Article
Functional Coating Effects of Silver Diamine Fluoride (SDF) on Artificial Caries Lesions: A Microhardness-Based Evaluation
by Mohammed H. Alshamrani, Reem A. Alajlan, Waad E. Alsaadi, Amjad M. Alabdulmohsen, Munira Abuthnain, Carlos Fernando Mourão and Adam Lowenstein
Materials 2025, 18(16), 3889; https://doi.org/10.3390/ma18163889 - 20 Aug 2025
Viewed by 189
Abstract
Background: Dental caries is a prevalent dental problem affecting primary and permanent teeth. Early demineralization of enamel lesions can be reversed through remineralization. Many studies have focused on caries prevention and disease progression arrest using silver diamine fluoride (SDF). No in vitro [...] Read more.
Background: Dental caries is a prevalent dental problem affecting primary and permanent teeth. Early demineralization of enamel lesions can be reversed through remineralization. Many studies have focused on caries prevention and disease progression arrest using silver diamine fluoride (SDF). No in vitro studies have compared the remineralization effects of different 38% SDF solutions on artificially demineralized enamel lesions. This study aimed to compare the remineralization potential of three commercial 38% silver diamine fluoride formulations on artificial enamel lesions in primary teeth using a pH cycling model. The hypothesis was as follows: different commercial SDF formulations would exhibit varying remineralization effects, as measured by surface microhardness, due to potential differences in their compositions. Materials and Methods: In this study, 75 primary molars were randomized into five groups (N = 15): I: baseline, II: SDF Riva Star Aqua® 38%, III: Riva Star® 38%, IV: SDF Advantage Arrest® 38%, and V: control. Artificial caries were created by submerging teeth in 10 mL of demineralization solution (pH 4.5) for three days in a light-resistant container, ensuring distinct visual changes in the enamel as per the International Caries Detection and Assessment System (ICDAS level 2). After pH cycling, all samples underwent a standardized Vickers microhardness test (VMHT) with a 50 g load for 15 s. Data were analyzed using one-way ANOVA and Tukey’s post hoc test, with a significance level set at p ≤ 0.05. Results: The one-way ANOVA test indicated a significant difference in microhardness among the groups (SDF Riva Star Aqua, SDF Riva Star, and SDF Advantage Arrest), with an F-value of 167.73 and p < 0.001. The post hoc Scheffé test showed that SDF Riva Star Aqua and SDF Riva Star were not significantly different (p = 0.388). However, SDF Advantage Arrest had a significantly higher mean microhardness compared to both groups (p < 0.001). Overall, these results show that SDF Advantage Arrest leads to greater microhardness than SDF Riva Star Aqua or SDF Riva Star. Conclusions: SDF Advantage Arrest showed superior performance among the SDF-treated groups, significantly increasing microhardness compared to SDF Riva Star Aqua and SDF Riva Star. This suggests that SDF Advantage Arrest offers enhanced remineralization and structural strengthening, making it the most effective option for managing demineralized primary teeth. Future research should investigate the long-term performance and mechanisms of these treatments to optimize clinical protocols for preserving primary tooth integrity. Full article
(This article belongs to the Section Biomaterials)
Show Figures

Figure 1

21 pages, 967 KiB  
Review
Recent Advances in the Application of Cucurbitacin B as an Anticancer Agent
by Dongge Yin, Hongyue Chen, Shuting Lin, Yufei Sun, Xiaohong Jing, Rongrong Chang, Yang Feng, Xiaoxv Dong, Changhai Qu, Jian Ni and Xingbin Yin
Int. J. Mol. Sci. 2025, 26(16), 8003; https://doi.org/10.3390/ijms26168003 - 19 Aug 2025
Viewed by 296
Abstract
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and [...] Read more.
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and primary liver cancer. CuB exerts anticancer effects through multiple mechanisms: inducing apoptosis, cell cycle arrest (G2/M or S phase), autophagy, and cytoskeleton disruption; inhibiting migration, invasion, and angiogenesis (via VEGF/FAK/MMP-9 and Wnt/β-catenin pathways); regulating metabolic reprogramming and immune responses; inducing pyroptosis, ferroptosis, and epigenetic changes; and reversing tumor drug resistance. These effects are associated with signaling pathways like JAK/STAT, PI3K/Akt/mTOR, and FOXM1-KIF20A. To improve its application potential, strategies such as structural modification (e.g., NO donor conjugation), combination therapy (with gemcitabine or cisplatin), and nanomaterial-based delivery (e.g., liposomes and exosome-mimicking nanoparticles) have been developed to enhance efficacy, reduce toxicity, and improve bioavailability. CuB shows broad-spectrum anticancer activity, but further research is needed to clarify the mechanisms underlying its cell-specific sensitivity and interactions with the immune system. This review systematically summarizes the physicochemical properties, anticancer mechanisms, and strategies for applying CuB and suggests future research directions, providing references for scientific research and clinical translation. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Graphical abstract

16 pages, 9092 KiB  
Article
Chromatin Remodeler RSF1 as an Oncogenic Driver and Therapeutic Target in Esophageal Squamous Cell Carcinoma
by Zhenhua Du, Zhili Jia, Yao Lin, Xudong Zhao, Gengsheng Cao and Hengbin Wang
Cells 2025, 14(16), 1262; https://doi.org/10.3390/cells14161262 - 15 Aug 2025
Viewed by 272
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent malignancy, ranking eleventh in incidence and seventh in mortality globally. Remodeling and Spacing factor 1 (RSF1), a chromatin remodeling factor, is frequently overexpressed in various tumors and correlates with poor prognosis. This study, combining public [...] Read more.
Esophageal squamous cell carcinoma (ESCC) is a prevalent malignancy, ranking eleventh in incidence and seventh in mortality globally. Remodeling and Spacing factor 1 (RSF1), a chromatin remodeling factor, is frequently overexpressed in various tumors and correlates with poor prognosis. This study, combining public database analysis and clinical sample validation, reveals significantly elevated RSF1 expression in ESCC tumor tissues, confirmed further in an ESCC orthotopic model. Functional assays show that RSF1 knockout (KO) significantly inhibits ESCC cell proliferation, migration, invasion, and in vivo tumor growth, while reintroducing RSF1 restores its oncogenic effects. Proteomic analysis highlights that RSF1 KO disrupts pathways associated with cell cycle control, apoptosis, and focal adhesion. Experimentally, RSF1 KO induces apoptosis and G2/M arrest, establishing its essential role in ESCC progression. Collectively, these findings establish RSF1 as an oncogenic driver and a promising therapeutic target in ESCC. Full article
Show Figures

Figure 1

33 pages, 10768 KiB  
Article
Pharmacodynamic-Driven Sequence-Dependent Synergy Effects in Pemetrexed-Osimertinib Combination Against Non-Small Cell Lung Cancer (NSCLC): Optimizing Synergy Through Sequential Interval
by Kuan Hu, Yu Xia, Tong Yuan, Yan Lin and Jin Yang
Pharmaceutics 2025, 17(8), 1044; https://doi.org/10.3390/pharmaceutics17081044 - 12 Aug 2025
Viewed by 409
Abstract
Background: Combining pemetrexed (PEM) with Osimertinib (OSI) improves outcomes in epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC), but optimal scheduling remains undefined. Sequential PEM → OSI strategies may outperform concurrent administration; however, the critical dosing interval determining synergy has not [...] Read more.
Background: Combining pemetrexed (PEM) with Osimertinib (OSI) improves outcomes in epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC), but optimal scheduling remains undefined. Sequential PEM → OSI strategies may outperform concurrent administration; however, the critical dosing interval determining synergy has not been explored. Methods: PEM pharmacodynamics were divided into an OSI-antagonized early phase (S-phase arrest and DNA damage accumulation) and OSI-synergized late phase (DNA damage peak, apoptosis initiation, and feedback EGFR activation). Time-course profiling of cell cycle, DNA damage, apoptosis, and EGFR pathways was evaluated under monotherapy or sequential combination regimens to elucidate the mechanisms underlying synergistic/antagonistic effects. Results: OSI antagonizes PEM’s early phase via G1 arrest but potently enhances late-phase apoptosis through Rad51/thymidylate synthase suppression, Bim upregulation, and inhibition of EGFR signaling. The 48 h interval PEM → OSI uniquely enabled complete early-phase execution and aligned OSI exposure with late-phase initiation, yielding robust synergy across OSI-sensitive cell lines. In contrast, the 24 h interval PEM → OSI sequence demonstrated synergy only in PEM-sensitive PC9 cells. Both concurrent PEM + OSI and OSI → PEM sequence induced attenuated DNA damage and apoptotic signaling. Conclusions: The 48 h interval PEM → OSI sequence maximizes efficacy by temporally segregating antagonistic and synergistic interactions. This pharmacodynamically optimized regimen represents a promising strategy for clinical translation. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Graphical abstract

15 pages, 3609 KiB  
Article
Activation of G Protein-Coupled Estrogen Receptor Induces p53 and ADAMTS1 to Inhibit Tumor Growth and Suppress Liver Cancer Metastasis
by Hee Jung Kwon, Ga Seul Lee, Jeong Hee Moon and Joohee Jung
Cancers 2025, 17(16), 2623; https://doi.org/10.3390/cancers17162623 - 11 Aug 2025
Viewed by 322
Abstract
Background/Objectives: Liver cancer is a common cause of cancer-related deaths among men and women globally. A disintegrin and metalloproteinase with thrombospondin motif 1 (ADAMST1) has been associated with various cancers, including prostate, esophageal, renal, and breast cancers. However, its role in liver [...] Read more.
Background/Objectives: Liver cancer is a common cause of cancer-related deaths among men and women globally. A disintegrin and metalloproteinase with thrombospondin motif 1 (ADAMST1) has been associated with various cancers, including prostate, esophageal, renal, and breast cancers. However, its role in liver cancer remains unclear. The aim of this study was to investigate the relationship between G protein-coupled estrogen (GPER) activation via its agonist, G1, and ADAMTS1 in suppressing liver cancer metastasis. Methods: Following preliminary assessment of Hep3B, Huh7, and SK-Hep-1 cells, SK-Hep-1 cells were selected owing to their elevated GPER expression and reduced cell viability. Cells were subjected to flow cytometry, RNA sequencing, and proteomics analyses. We established an SK-Hep-1 xenograft model for in vivo analysis. Results: We observed G1-induced G2-M phase cell cycle arrest, increased p53 and p21, and decreased cell cycle-related factors. In vivo, G1 significantly inhibited tumor growth and increased p53 protein expression. ADAMTS1, a metastasis regulator, was significantly upregulated by G1. G1 reduced the proliferating cell nuclear antigen and increased E-cadherin expression in SK-Hep-1 cells and in vivo. Tumor invasion was reduced with G1 and ADAMTS1 expression. In vivo, G1 reduced liver metastasis, increased E-cadherin, and decreased vimentin and proliferating cell nuclear antigen in primary tumor tissues and increased ADAMTS1 at the tumor edge. Conclusions: GPER agonists, such as G1, show potential for suppressing liver cancer progression and metastasis. Full article
(This article belongs to the Special Issue Liver Cancer: Improving Standard Diagnosis and Therapy: 2nd Edition)
Show Figures

Figure 1

16 pages, 3848 KiB  
Article
Reversing Preeclampsia Pathology: AXL Inhibition Restores Mitochondrial Function and ECM Balance
by Archarlie Chou, Benjamin Davidson, Paul R. Reynolds, Brett E. Pickett and Juan A. Arroyo
Cells 2025, 14(16), 1229; https://doi.org/10.3390/cells14161229 - 8 Aug 2025
Viewed by 269
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal morbidity that affects 2–8% of pregnancies worldwide, driven by placental dysfunction and systemic inflammation. Growth arrest-specific protein 6 (Gas6) and its receptor AXL play pivotal roles in PE pathogenesis, promoting trophoblast impairment and [...] Read more.
Preeclampsia (PE) is a leading cause of maternal and fetal morbidity that affects 2–8% of pregnancies worldwide, driven by placental dysfunction and systemic inflammation. Growth arrest-specific protein 6 (Gas6) and its receptor AXL play pivotal roles in PE pathogenesis, promoting trophoblast impairment and vascular dysregulation. This study investigated the transcriptomic reversal effects of AXL Receptor Tyrosine Kinase (AXL) inhibition in a Gas6-induced rat model of PE using RNA sequencing (RNA-seq). Pregnant rats were administered Gas6 to induce PE-like symptoms such as hypertension and proteinuria; a subset also received the AXL inhibitor R428. RNA-seq of placental tissues revealed 2331 differentially expressed genes (DEGs) in Gas6-AXLi versus Gas6 (1277 upregulated, 1054 downregulated). Protein–protein interaction networks and Gene Ontology enrichment highlighted upregulated mitochondrial functions, including electron transport chain components (e.g., NDUFC2, COX5A), suggesting enhanced energy metabolism. In the secondary analysis that compared Gas6 to Control, Gas6-upregulated extracellular matrix proteins (e.g., COL4A1, LAMC1) linked to fibrosis were reversed by AXL inhibition, indicating ameliorated placental remodeling. AXL inhibition activated compensatory pathways beyond Gas6 blockade, unveiling novel mechanisms for PE resolution. These findings position AXL inhibitors as promising therapeutics, offering insights into mitochondrial and fibrotic targets to mitigate this enigmatic disorder. Full article
Show Figures

Figure 1

18 pages, 5890 KiB  
Article
Targeting Hippo Signaling Pathway with a Boron Derivative, Sodium Pentaborate Pentahydrate (NaB): Therapeutic Strategies in Colorectal Cancer
by Büşra Yüksel, Fikrettin Şahin and Nezaket Türkel
Pharmaceuticals 2025, 18(8), 1171; https://doi.org/10.3390/ph18081171 - 8 Aug 2025
Viewed by 308
Abstract
Background/Objectives: Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, highlighting the urgent need for novel therapeutic strategies. This study aimed to investigate the anticancer potential of sodium pentaborate pentahydrate (NaB) in CRC by evaluating its effects on human colorectal [...] Read more.
Background/Objectives: Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, highlighting the urgent need for novel therapeutic strategies. This study aimed to investigate the anticancer potential of sodium pentaborate pentahydrate (NaB) in CRC by evaluating its effects on human colorectal cancer cell lines and elucidating underlying molecular mechanisms. Methods: The cytotoxic and molecular effects of NaB were assessed in three human CRC cell lines (HCT-116, HT-29, and COLO-205) and one normal colon epithelial cell line (CCD-18CO). Cell viability assays were conducted to determine time- and dose-dependent responses. Apoptosis, cell cycle progression, colony formation, and migration capacity were evaluated. Gene and protein expression analyses were performed to examine apoptosis-related, DNA damage response, cell cycle, and Hippo signaling pathway components. Results: NaB significantly reduced cancer cell viability in a time- and dose-dependent manner, with minimal cytotoxicity to normal colon cells. It induced marked apoptosis, especially in HCT-116 and COLO-205 cells, and caused G2/M cell cycle arrest. In HCT-116 cells, NaB suppressed proliferation by downregulating PCNA and MKI-67 and reduced colony formation and migration. Molecular analyses revealed upregulation of pro-apoptotic BAX and downregulation of BCL-2, ATM, ATR, and cell cycle–related genes. NaB also inhibited oncogenic Hippo signaling by enhancing YAP1 phosphorylation and decreasing CTGF and CYR61 expression. Conclusions: These findings demonstrate that sodium pentaborate pentahydrate exerts selective anticancer effects on colorectal cancer cells through the induction of apoptosis, cell cycle arrest, and suppression of key oncogenic pathways. NaB represents a promising candidate for further development as a therapeutic agent in CRC treatment. Full article
Show Figures

Graphical abstract

19 pages, 3228 KiB  
Article
N-Degron-Based PROTAC Targeting PLK1: A Potential Therapeutic Strategy for Cervical Cancer
by Pethaiah Gunasekaran, Sang Chul Shin, Yeon Sil Hwang, Jihyeon Lee, Yeo Kyung La, Min Su Yim, Hak Nam Kim, Tae Wan Kim, Eunjung Yang, Soo Jae Lee, Jung Min Yoon, Eunice EunKyeong Kim, Seob Jeon, Eun Kyoung Ryu and Jeong Kyu Bang
Pharmaceutics 2025, 17(8), 1027; https://doi.org/10.3390/pharmaceutics17081027 - 7 Aug 2025
Viewed by 455
Abstract
Background: Cervical cancer remains a major global health concern, with existing chemotherapy facing limited effectiveness owing to resistance. Polo-like kinase 1 (PLK1) overexpression in cervical cancer cells is a promising target for developing novel therapies to overcome chemoresistance and improve treatment efficacy. [...] Read more.
Background: Cervical cancer remains a major global health concern, with existing chemotherapy facing limited effectiveness owing to resistance. Polo-like kinase 1 (PLK1) overexpression in cervical cancer cells is a promising target for developing novel therapies to overcome chemoresistance and improve treatment efficacy. Methods: In this study, we developed a novel PROTAC, NC1, targeting PLK1 PBD via the N-end rule pathway. Results: This PROTAC effectively depleted the PLK1 protein in HeLa cells by inducing protein degradation. The crystal structure of the PBD-NC1 complex identified key PLK1 PBD binding interactions and isothermal titration calorimetry (ITC) confirmed a binding affinity of 6.06 µM between NC1 and PLK1 PBD. NC1 significantly decreased cell viability with an IC50 of 5.23 µM, induced G2/M phase arrest, and triggered apoptosis in HeLa cells. In vivo, NC1 suppressed tumor growth in a HeLa xenograft mouse model. Conclusions: This research highlights the potential of N-degron-based PROTACs targeting the PLK1 protein in cancer therapies, highlighting their potential in future cervical anticancer treatment strategies. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

22 pages, 28302 KiB  
Article
IGF2BP3 as a Novel Prognostic Biomarker and Therapeutic Target in Lung Adenocarcinoma
by Feiming Hu, Chenchen Hu, Yuanli He, Lin Guo, Yuanjie Sun, Chenying Han, Xiyang Zhang, Junyi Ren, Jinduo Han, Jing Wang, Junqi Zhang, Yubo Sun, Sirui Cai, Dongbo Jiang, Kun Yang and Shuya Yang
Cells 2025, 14(15), 1222; https://doi.org/10.3390/cells14151222 - 7 Aug 2025
Viewed by 413
Abstract
RNA-binding proteins (RBPs), particularly IGF2BP3, play critical but underexplored roles in lung adenocarcinoma (LUAD). This study investigated IGF2BP3′s clinical and functional significance using single-cell/RNA sequencing, validated by qPCR, Western blot, and immunohistochemistry. The results show IGF2BP3 was significantly upregulated in LUAD tissues and [...] Read more.
RNA-binding proteins (RBPs), particularly IGF2BP3, play critical but underexplored roles in lung adenocarcinoma (LUAD). This study investigated IGF2BP3′s clinical and functional significance using single-cell/RNA sequencing, validated by qPCR, Western blot, and immunohistochemistry. The results show IGF2BP3 was significantly upregulated in LUAD tissues and associated with advanced-stage, larger tumors, lymph node metastasis, and poor prognosis. A prognostic nomogram confirmed its independent predictive value. Functionally, IGF2BP3 knockdown suppressed proliferation, and induced G2/M arrest and apoptosis. GSEA linked high IGF2BP3 to cell cycle activation and low expression to metabolic pathways. Notably, high IGF2BP3 correlated with immune evasion markers (downregulated CD4+ effector T cells, upregulated Th2 cells), while TIDE analysis suggested a better immunotherapy response in low-expressing patients. Drug screening identified BI-2536 as a potential therapy for low-IGF2BP3 cases, supported by strong molecular docking affinity (−7.55 kcal/mol). These findings establish IGF2BP3 as a key driver of LUAD progression and a promising target for immunotherapy and precision medicine. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

19 pages, 3082 KiB  
Article
Anticancer Activity of Vitex agnus-castus Seed Extract on Gastric Cancer Cells
by Özlem Türksoy-Terzioğlu, Feyza Tosya, Ayşe Büşranur Çelik, Sibel Bölek, Levent Gülüm, Gökhan Terzioğlu and Yusuf Tutar
Nutrients 2025, 17(15), 2564; https://doi.org/10.3390/nu17152564 - 6 Aug 2025
Viewed by 360
Abstract
Background/Objectives: Vitex agnus-castus has been traditionally used to treat hormonal disorders, and recent evidence suggests its potential anticancer properties. However, its effects on gastric cancer remain unclear. Methods: This study examined the cytotoxic, apoptotic, and anti-metastatic effects of hydroalcoholic Vitex agnus-castus [...] Read more.
Background/Objectives: Vitex agnus-castus has been traditionally used to treat hormonal disorders, and recent evidence suggests its potential anticancer properties. However, its effects on gastric cancer remain unclear. Methods: This study examined the cytotoxic, apoptotic, and anti-metastatic effects of hydroalcoholic Vitex agnus-castus seed extract in gastric cancer cells. Antioxidant capacity (DPPH, ABTS) and total phenolic and flavonoid contents were analyzed. Cytotoxicity was assessed using the MTT assay in HGC27, MKN45, and AGS gastric cancer cell lines and CCD-1072Sk fibroblasts. Apoptosis, mitochondrial membrane potential (MMP), and cell cycle changes were evaluated via Annexin V-FITC/PI, Rhodamine 123, and PI staining, respectively. RT-qPCR and gene enrichment analyses were conducted to investigate the molecular mechanisms. Apoptosis-related protein expression was analyzed through enzyme-linked immunosorbent assay (ELISA). Results: The extract exhibited high antioxidant activity and a significant phenolic and flavonoid content. It reduced cell viability in a dose-dependent manner in gastric cancer cells, while exerting low toxicity in fibroblasts. It significantly increased apoptosis, induced G0/G1-phase cell cycle arrest, upregulated pro-apoptotic genes (CASP3, CASP7, TP53, BCL2L11), and downregulated anti-apoptotic genes (XIAP, NOL3). Gene enrichment analysis highlighted pathways like apoptosis, necrosis, and cysteine endopeptidase activity. The extract also disrupted MMP, inhibited migration and spheroid formation, suppressed EMT markers (SNAIL, SLUG, TWIST1, N-CADHERIN), and upregulated E-CADHERIN. The expression of Caspase 3 and Bax proteins increased and Bcl2 protein decreased. Conclusions: These findings suggest that Vitex agnus-castus seed extract exerts strong anticancer effects in gastric cancer cells by promoting apoptosis, reducing proliferation, and inhibiting migration. Further studies are warranted to explore its clinical relevance. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Graphical abstract

17 pages, 2353 KiB  
Article
Repurposing a Lipid-Lowering Agent to Inhibit TNBC Growth Through Cell Cycle Arrest
by Yi-Chiang Hsu, Kuan-Ting Lee, Sung-Nan Pei, Kun-Ming Rau and Tai-Hsin Tsai
Curr. Issues Mol. Biol. 2025, 47(8), 622; https://doi.org/10.3390/cimb47080622 - 5 Aug 2025
Viewed by 310
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and therapeutically challenging subtype of breast cancer due to its lack of estrogen receptors, progesterone receptors, and HER2 (Human epidermal growth factor receptor 2) expression, which severely limits available treatment options. Recently, Simvastatin—a widely used [...] Read more.
Triple-negative breast cancer (TNBC) is a highly aggressive and therapeutically challenging subtype of breast cancer due to its lack of estrogen receptors, progesterone receptors, and HER2 (Human epidermal growth factor receptor 2) expression, which severely limits available treatment options. Recently, Simvastatin—a widely used HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase inhibitor for hyperlipidemia—has garnered interest for its potential anticancer effects. This study investigates the therapeutic potential of Simvastatin in triple-negative breast cancer (TNBC). The results demonstrate that Simvastatin significantly inhibits the proliferation of TNBC cells, particularly MDA-MB-231, in a dose- and time-dependent manner. Mechanistically, Simvastatin primarily induces G1 phase cell cycle arrest to exert its antiproliferative effects, with no significant evidence of apoptosis or necrosis. These findings support the potential repositioning of Simvastatin as a therapeutic agent to suppress TNBC cell growth. Further analysis shows that Simvastatin downregulates cyclin-dependent kinase 4 (CDK4), a key regulator of the G1/S cell cycle transition and a known marker of poor prognosis in breast cancer. These findings highlight a novel, apoptosis-independent mechanism of Simvastatin’s anticancer action in TNBC. Importantly, given that many breast cancer patients also suffer from hyperlipidemia, Simvastatin offers dual therapeutic benefits—managing both lipid metabolism and tumor cell proliferation. Thus, Simvastatin holds promise as an adjunctive therapy in the treatment of TNBC and warrants further clinical investigation. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

18 pages, 3940 KiB  
Article
CTCF Represses CIB2 to Balance Proliferation and Differentiation of Goat Myogenic Satellite Cells via Integrin α7β1–PI3K/AKT Axis
by Changliang Gong, Huihui Song, Zhuohang Hao, Zhengyi Zhang, Nanjian Luo and Xiaochuan Chen
Cells 2025, 14(15), 1199; https://doi.org/10.3390/cells14151199 - 5 Aug 2025
Viewed by 616
Abstract
Skeletal muscle development is a critical economic trait in livestock, governed by myogenic satellite cell regulation. Integrins mediate mechanical anchorage to the ECM and enable ECM–intracellular signaling. CIB2, as an EF-hand-domain protein involved in mechanotransduction, shows significant developmental regulation in goat muscle. [...] Read more.
Skeletal muscle development is a critical economic trait in livestock, governed by myogenic satellite cell regulation. Integrins mediate mechanical anchorage to the ECM and enable ECM–intracellular signaling. CIB2, as an EF-hand-domain protein involved in mechanotransduction, shows significant developmental regulation in goat muscle. Although the role of CIB2 in skeletal muscle growth is poorly characterized, we observed pronounced developmental upregulation of IB2 in postnatal goat muscle. CIB2 expression increased >20-fold by postnatal day 90 (P90) compared to P1, sustaining elevation through P180 (p < 0.05). Functional investigations indicated that siRNA-mediated knockdown of CIB2 could inhibit myoblast proliferation by inducing S-phase arrest (p < 0.05) and downregulating the expression of CDK4/Cyclin D/E. Simultaneously, CIB2 interference treatment was found to decrease the proliferative activity of goat myogenic satellite cells, yet it significantly promoted differentiation by upregulating the expression of MyoD/MyoG/MyHC (p < 0.01). Mechanistically, CTCF was identified as a transcriptional repressor binding to an intragenic region of the CIB2 gene locus (ChIP enrichment: 2.3-fold, p < 0.05). Knockdown of CTCF induced upregulation of CIB2 (p < 0.05). RNA-seq analysis established CIB2 as a calcium signaling hub: its interference activated IL-17/TNF and complement cascades, while overexpression suppressed focal adhesion/ECM–receptor interactions and enriched neuroendocrine pathways. Collectively, this study identifies the CTCF-CIB2–integrin α7β1–PI3K/AKT axis as a novel molecular mechanism that regulates the balance of myogenic fate in goats. These findings offer promising targets for genomic selection and precision breeding strategies aimed at enhancing muscle productivity in ruminants. Full article
Show Figures

Figure 1

24 pages, 5287 KiB  
Article
Influence of Sample Mass and Pouring Temperature on the Effectiveness of Thermal Analysis for Estimating Gray Iron Inoculation Potential
by Raymundo del Campo-Castro, Manuel Castro-Román, Edgar-Ivan Castro-Cedeno and Martín Herrera-Trejo
Materials 2025, 18(15), 3640; https://doi.org/10.3390/ma18153640 - 2 Aug 2025
Viewed by 286
Abstract
Thermal analysis (TA) has been a valuable tool for controlling the carbon equivalent (CE) of cast irons. Additionally, this technique can provide enhanced control over melt quality, allowing for the avoidance of defects such as undesirable graphite morphology and the formation of carbides. [...] Read more.
Thermal analysis (TA) has been a valuable tool for controlling the carbon equivalent (CE) of cast irons. Additionally, this technique can provide enhanced control over melt quality, allowing for the avoidance of defects such as undesirable graphite morphology and the formation of carbides. To obtain the most valuable information from the TA, it is necessary to minimize the variations in the filling operation of the TA cups. However, the mass and pouring temperature of TA cups can vary in TA’s typical foundry operations. A design of experiments was performed to determine whether specific parameters of cooling curves used for quality control can distinguish the inoculation effect in the melt when the mass and the pouring temperature of TA cups are varied. The minimum temperature of the eutectic arrest proved to be a robust inoculation potential control parameter when variations in the cup’s mass were within a range of 268–390 g and were filled at any pouring temperature between 1235 and 1369 °C. Lighter cups under 268 g and poured at a low temperature are not suitable for controlling inoculation potential by TA; however, they remain helpful in controlling CE. These later cups are related to cooling times of less than 180 s, which can serve as a criterion for discarding unsuitable samples. A bimodal population of cell surfaces was revealed in the samples, with the population of small cells being proportionally more numerous in samples with lower TEmin values. Full article
Show Figures

Figure 1

Back to TopTop