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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,321)

Search Parameters:
Keywords = Epithelial to mesenchymal transition (EMT)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5023 KB  
Article
Integrating Network Pharmacology, Machine Learning, and Experimental Validation to Elucidate the Mechanism of Cardamonin in Treating Idiopathic Pulmonary Fibrosis
by Wenyue Zhang, Yi Guo, Qiushi Wang, Kai Wang, Huning Zhang, Sirong Chang, Anning Yang, Zhihong Liu and Yue Sun
Int. J. Mol. Sci. 2026, 27(1), 249; https://doi.org/10.3390/ijms27010249 - 25 Dec 2025
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and irreversible interstitial lung disease characterized by progressive scarring of the lungs. The available therapeutic strategies are limited and primarily focus on slowing disease progression rather than achieving fibrosis reversal. Cardamonin (CDN), a food-derived natural chalcone, [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic and irreversible interstitial lung disease characterized by progressive scarring of the lungs. The available therapeutic strategies are limited and primarily focus on slowing disease progression rather than achieving fibrosis reversal. Cardamonin (CDN), a food-derived natural chalcone, has exhibited anti-fibrotic activity in liver and kidney fibrosis models; however, its role and underlying mechanism in IPF remain unelucidated. Herein, we integrated network pharmacology, machine learning, molecular simulations, and in vitro experiments. Network pharmacology identified 135 overlapping targets between CDN and IPF, which demonstrated a significant enrichment in the Phosphatidylinositol 3-Kinase/Protein Kinase B signaling pathway (PI3K/AKT). Machine learning further prioritized 6 core targets, with IGF1 emerging as a key candidate. Molecular docking revealed a favorable binding energy of −7.9 kcal/mol for the CDN-IGF1 complex. Subsequent 100 ns molecular dynamics simulations further confirmed its robust binding stability, yielding a mean binding free energy of −150.978 kcal/mol. In vitro, CDN significantly mitigated fibrosis in bleomycin (BLM)-challenged A549 cells, downregulating the expression of α-smooth muscle actin (α-SMA) and fibronectin. This effect was accompanied by a beneficial reversal of epithelial–mesenchymal transition (EMT), as indicated by increased E-cadherin levels and decreased vimentin expression. Mechanistically, CDN significantly suppressed the IGF1/PI3K/AKT axis; this inhibitory effect was partially reversed by exogenous IGF1 supplementation and further enhanced by the PI3K-specific inhibitor LY294002. This work provides the evidence that CDN alleviates BLM-induced pulmonary fibrosis by targeting the IGF1/PI3K/AKT-EMT axis. These findings lend support to a robust mechanistic basis for developing CDN as a potential therapeutic candidate for IPF. It should be noted that these conclusions are drawn from in vitro experiments using A549 cells, and further validation in primary alveolar epithelial cells and animal models is warranted to confirm their physiological relevance. Full article
(This article belongs to the Section Molecular Pharmacology)
13 pages, 3394 KB  
Article
Resveratrol Prevents Breast Cancer Metastasis by Inhibiting Wnt/β-Catenin Pathway-Mediated Epithelial–Mesenchymal Transition
by Xue Fang, En Ma, Runshu Wang, Jingwei Zhang, Yirong Tang, Jinxiao Chen, Weidong Zhu, Da Wo and Dan-ni Ren
Pharmaceuticals 2026, 19(1), 41; https://doi.org/10.3390/ph19010041 - 23 Dec 2025
Abstract
Background: Breast cancer is the most prevalent cancer in women, and metastatic breast cancer remains a major cause of cancer-related deaths. Resveratrol (RSV) is a natural compound found in various plants and is known to exhibit various anti-cancer effects. The present study [...] Read more.
Background: Breast cancer is the most prevalent cancer in women, and metastatic breast cancer remains a major cause of cancer-related deaths. Resveratrol (RSV) is a natural compound found in various plants and is known to exhibit various anti-cancer effects. The present study aims to investigate the therapeutic effects and mechanisms of RSV in inhibiting breast cancer metastasis in a murine model of 4T1 breast tumor that shares close molecular features with human triple negative breast cancer. Methods: Murine breast cancer 4T1 cells were used to examine the effects of RSV on breast cancer metastasis and epithelial–mesenchymal transition (EMT). In vitro cell proliferation and Transwell migration assays and in vivo 4T1 tumor transplantation models were established in female Balb/c mice to determine the anti-metastatic effects of RSV and its mechanism of action. Results: RSV significantly inhibited 4T1 tumor cell migration and significantly decreased expression levels of EMT markers Snail and Vimentin, as well as the nuclear translocation of β-catenin both in vitro and in vivo. Knockdown of β-catenin similarly reduced the expression levels of EMT markers. RSV significantly decreased the number of lung metastases in 4T1-implanted mice by inhibiting Wnt/β-catenin signaling pathway activation. RSV (150 mg/kg/day) reduced the number of visible tumor metastatic nodules and the histological count of metastatic lung carcinomas by 51.82% and 62.58%, respectively, compared to vehicle administration. Conclusions: Our study provides important new mechanistic insight into the strong anti-cancer effects of RSV in inhibiting 4T1 breast cancer metastasis by preventing Wnt/β-catenin signaling pathway-mediated epithelial–mesenchymal transition. These findings suggest the therapeutic potential of RSV as a promising drug in the treatment of metastatic breast cancer. Full article
(This article belongs to the Section Medicinal Chemistry)
20 pages, 4121 KB  
Article
Targeting Growth Hormone Receptor to Overcome Therapy Resistance in Non-Small Cell Lung Cancer
by Arshad Ahmad, Reetobrata Basu, Caden Fyffe, Reece Geiger, Christopher Walsh, Delany Minto, Edward Brenya, Amrutha Varshini Alur, Sebastian J. C. M. M. Neggers and John J. Kopchick
Int. J. Mol. Sci. 2026, 27(1), 115; https://doi.org/10.3390/ijms27010115 - 22 Dec 2025
Viewed by 83
Abstract
Lung cancer (LC) remains the leading cause of cancer-related death in the United States despite advances in therapy. Growth hormone (GH) action has been implicated in tumor progression and therapy resistance across multiple cancers, but its role in LC, particularly non-small cell lung [...] Read more.
Lung cancer (LC) remains the leading cause of cancer-related death in the United States despite advances in therapy. Growth hormone (GH) action has been implicated in tumor progression and therapy resistance across multiple cancers, but its role in LC, particularly non-small cell lung cancer (NSCLC), remains poorly defined. In cancer cells, GH promotes chemoresistance through upregulation of drug-efflux pumps, induction of epithelial-to-mesenchymal transition (EMT), and inhibition of apoptosis. Notably, GH receptor (GHR) expression is significantly elevated in NSCLC compared to normal lung tissue, suggesting a potential therapeutic opportunity. In this study, we investigated the impact of GH action on therapy resistance and tumor progression using integrated transcriptomic analyses and in vitro experiments. Analyses of transcriptomic data from NSCLC patients revealed that high tumoral GHR expression correlates with reduced overall survival, and with upregulation of genes involved in distinct therapy refractory pathways. Our in vitro studies demonstrated that GH promotes chemoresistance in NSCLC cell lines through activation of ABC transporters and EMT pathways, whereas GHR antagonism with the GH receptor antagonist, pegvisomant, effectively counteracts these effects and improves chemotherapy efficacy significantly. Together, our findings identify GHR signaling as a contributor to aggressive and therapy-resistant phenotypes in NSCLC in vitro and suggest that GHR antagonism may enhance chemotherapy sensitivity. These results provide a rationale for further in vivo and mechanistic studies to evaluate the therapeutic potential of targeting GHR in NSCLC. Full article
Show Figures

Figure 1

20 pages, 2031 KB  
Review
GalNAc-Transferases in Cancer
by Shruthi C. Iyer, Dinesh Kumar Srinivasan and Rajeev Parameswaran
Biomedicines 2026, 14(1), 5; https://doi.org/10.3390/biomedicines14010005 - 19 Dec 2025
Viewed by 200
Abstract
Background/Objectives: The polypeptide N-acetylgalactosaminyltransferase (GALNT) family initiates mucin-type O-glycosylation, a post-translational modification that plays a pivotal role in cellular signaling, adhesion, and immune evasion. Dysregulated GALNT expression has been increasingly implicated in carcinogenesis. Methods: We reviewed the literature on the [...] Read more.
Background/Objectives: The polypeptide N-acetylgalactosaminyltransferase (GALNT) family initiates mucin-type O-glycosylation, a post-translational modification that plays a pivotal role in cellular signaling, adhesion, and immune evasion. Dysregulated GALNT expression has been increasingly implicated in carcinogenesis. Methods: We reviewed the literature on the expression, function, and clinical relevance of GALNT isoforms across various cancers, with a focus on their mechanistic roles, biomarker potential, and therapeutic implications. Results: Aberrant GALNT expression is observed in numerous malignancies, including breast, colorectal, gastric, lung, ovarian, and hepatocellular carcinomas. Isoforms such as GALNT1, -T2, -T3, and -T14 contribute to tumorigenesis by modulating the glycosylation of mucins such as Mucin-1 (MUC1), epithelial growth factor receptors (EGFR), and other signaling proteins. These alterations promote cancer cell proliferation, metastasis, epithelial–mesenchymal transition (EMT), and chemoresistance. Deranged GALNT expression is frequently associated with poor prognosis, and certain GALNT genotypes predict treatment response. However, functional redundancy among isoforms poses challenges for selective targeting. Conclusions: Despite their strong potential as modulators of cancer progression, GALNTs face substantial limitations in terms of substrate identification, mechanistic clarity, immune relevance, and therapeutic tractability. Overcoming these challenges requires advanced glycoproteomics, development of isoform-specific tools, and integrated studies across cancer and immunology to fully harness GALNT biology for clinical benefit. Full article
(This article belongs to the Special Issue Role of Glycomics in Health and Diseases)
Show Figures

Graphical abstract

30 pages, 2235 KB  
Review
Microplastics and Nanoplastics in Cancer Progression: Biology and Public Health
by Richard Ponce-Cusi, Claudia Barletta-Carrillo and Nesstor Pilco-Ferreto
Biomedicines 2026, 14(1), 1; https://doi.org/10.3390/biomedicines14010001 - 19 Dec 2025
Viewed by 326
Abstract
Microplastics and nanoplastics (MPs/NPs), emerging as pervasive environmental contaminants, have raised growing concern due to their potential implications for human health. Among their diverse biological effects, recent evidence highlights their capacity to cross biological barriers, accumulate in tissues, and interact with cellular components [...] Read more.
Microplastics and nanoplastics (MPs/NPs), emerging as pervasive environmental contaminants, have raised growing concern due to their potential implications for human health. Among their diverse biological effects, recent evidence highlights their capacity to cross biological barriers, accumulate in tissues, and interact with cellular components in ways that may promote carcinogenesis. MPs/NPs can cause oxidative stress, inflammation, and epithelial barrier dysfunction, leading to cellular homeostasis disruption. Their interaction with endothelial cells and immune components further exacerbates pro-tumorigenic processes, including angiogenesis, immune evasion, and epithelial–mesenchymal transition (EMT), thereby potentially facilitating tumor initiation and progression. At the cellular level, these particles are internalized through various endocytic pathways, where they are associated with oxidative stress, inflammation, DNA damage, and barrier dysfunction—processes that have been linked to carcinogenesis. This review synthesizes current evidence on the cellular and molecular mechanisms through which MPs/NPs may contribute to cancer development, with particular emphasis on their interactions with endothelial cells and the tumor microenvironment. It highlights the need for further mechanistic and epidemiological studies to clarify the potential role of these particles in carcinogenesis. Given the increasing global production and environmental ubiquity of plastic particles, understanding their direct contribution to cancer development is critical for advancing both public health strategies and environmental regulations. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

22 pages, 4207 KB  
Article
SS18::SSX and BRD9 Modulate Synovial Sarcoma Differentiation
by Anna Kuntze, Victor Banerjee, Marcel Trautmann, Charlotte Pünt, Ruth Berthold, Pascal Hauser, Lucas Scholl, Eva Wardelmann, Kornelius Kerl, Wolfgang Hartmann and Ilka Isfort
Cells 2025, 14(24), 2022; https://doi.org/10.3390/cells14242022 - 18 Dec 2025
Viewed by 198
Abstract
Synovial sarcoma (SySa) is a malignant soft tissue tumor that is characterized by an SS18::SSX fusion protein, which integrates into BAF chromatin remodeling complexes and alters global gene transcription. Despite its uniform genetic driver, SySa displays striking histomorphological and phenotypic heterogeneity, including spindle [...] Read more.
Synovial sarcoma (SySa) is a malignant soft tissue tumor that is characterized by an SS18::SSX fusion protein, which integrates into BAF chromatin remodeling complexes and alters global gene transcription. Despite its uniform genetic driver, SySa displays striking histomorphological and phenotypic heterogeneity, including spindle cell, glandular and poorly differentiated patterns. Prognosis is variable, with around 50% of patients developing metastases. Limited response to chemotherapy highlights the need for a better understanding of the underlying molecular mechanisms to guide alternative therapeutic strategies. Given the pivotal function of BAF complexes in SySa and their recently described impact on cellular differentiation processes, this study aims to investigate the role of SS18::SSX and specific BAF subunits in SySa differentiation. Nanostring analysis revealed that silencing of SS18::SSX and the GBAF subunit BRD9 modulates the cellular differentiation pathways. SS18::SSX and BRD9 were found to regulate epithelial–mesenchymal-transition (EMT)-associated factors of Snail and Slug on different levels, with SS18::SSX repressing E-Cadherin expression. Published single-cell RNA sequencing data were analyzed to validate our finding that BRD9 contributes to SySa EMT regulation. Our study provides novel insights into the multilayered regulation of key EMT players by SS18::SSX and BRD9 in SySa, thereby defining tumor phenotype and (potentially) prognosis. Full article
Show Figures

Figure 1

12 pages, 227 KB  
Review
Dynamic Interplay of Epithelial–Mesenchymal and Mesenchymal–Epithelial Transitions in Cochlear Development and Disease: Molecular Mechanisms and Therapeutic Implications
by Jingyi Zhu, Sihan Huang, Jifang Zhang, Tianyu Gong, Zhongyuan Fei, Penghui Chen, Shule Hou and Jun Yang
Biomedicines 2025, 13(12), 3115; https://doi.org/10.3390/biomedicines13123115 - 18 Dec 2025
Viewed by 203
Abstract
Epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET) are evolutionarily conserved cellular processes defined by reversible conversions between epithelial and mesenchymal phenotypes. As dynamic regulatory programs, they contribute to cell fate determination, tissue remodeling, and functional maturation during embryogenesis. In the cochlea, emerging evidence [...] Read more.
Epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET) are evolutionarily conserved cellular processes defined by reversible conversions between epithelial and mesenchymal phenotypes. As dynamic regulatory programs, they contribute to cell fate determination, tissue remodeling, and functional maturation during embryogenesis. In the cochlea, emerging evidence suggests that EMT/MET are implicated in certain aspects of sensory epithelium development. This review systematically dissects the molecular mechanisms underlying EMT and MET during cochlear development, along with the regulatory networks that control cell fate and signaling pathways. We further explore the emerging functions of these processes in cochlear pathologies, integrating recent advances to clarify their physiological and pathological relevance. By providing a comprehensive synthesis, this work aims to establish a theoretical framework for developing therapeutic strategies against related disorders. Full article
(This article belongs to the Section Molecular Genetics and Genetic Diseases)
28 pages, 3140 KB  
Review
The Impact of Senescence-Associated Secretory Phenotype (SASP) on Head and Neck Cancers: From Biology to Therapy
by Md Tanjim Alam, Mishfak A. M. Mansoor, Sarah A. Ashiqueali, Pawel Golusinski, Ewelina Golusinska-Kardach, Joanna K. Strzelczyk, Blazej Rubis, Wojciech Golusinski and Michal M. Masternak
Cancers 2025, 17(24), 4024; https://doi.org/10.3390/cancers17244024 - 17 Dec 2025
Viewed by 436
Abstract
Cellular senescence is defined as a state of permanent cell cycle arrest, providing a natural barrier against cancer. However, senescent cells are very metabolically active and secrete a complex mixture of bioactive molecules collectively known as the senescence-associated secretory phenotype (SASP), which play [...] Read more.
Cellular senescence is defined as a state of permanent cell cycle arrest, providing a natural barrier against cancer. However, senescent cells are very metabolically active and secrete a complex mixture of bioactive molecules collectively known as the senescence-associated secretory phenotype (SASP), which play a dual role in cancer biology. While the SASP can suppress tumors by facilitating immunosurveillance, it can also promote tumor progression by fostering a pro-inflammatory milieu, stimulating angiogenesis, enhancing invasiveness, and enabling immune evasion. In Head and Neck Cancers (HNCs), a highly heterogeneous group of malignancies, SASP has emerged as a critical player in disease progression and treatment resistance. Persistent DNA damage response (DDR) signaling drives SASP and thereby contributes to the progression of head and neck cancer by modulating the tumour microenvironment. It influences the tumor microenvironment (TME) by facilitating epithelial-to-mesenchymal transition (EMT), promoting cancer stem cell-like properties, and impairing the efficacy of radiotherapy, chemotherapy, and immune checkpoint inhibitors. These effects underscore the need for targeted interventions to regulate SASP activity. This review presents a comprehensive overview of the molecular mechanisms underlying SASP generation and its effects on HNCs. We discuss the dual roles of SASP in tumor suppression and progression, its contribution to therapy resistance, and emerging therapeutic strategies, including novel senolytic and senomorphic drugs. Finally, we highlight key challenges and future directions for translating SASP-targeted therapies into clinical practice, emphasizing the need for biomarker discovery, and a deeper understanding of SASP heterogeneity. By targeting the SASP, there is potential to enhance therapeutic outcomes and improve the management of HNCs. Full article
Show Figures

Figure 1

20 pages, 38977 KB  
Article
A Trifluoromethyl Quinazoline Compound Regulates the Epithelial–Mesenchymal Transition of Prostatic Hyperplasia Cells by Inhibiting the Secretion of TGF-β1 in Stromal Cells
by Lu Chen, Di Zhang, Gang Yu, Sha Cheng, Bixue Xu, Jia Yu, Jiming Liu and Heng Luo
Curr. Issues Mol. Biol. 2025, 47(12), 1057; https://doi.org/10.3390/cimb47121057 - 17 Dec 2025
Viewed by 172
Abstract
Benign prostatic hyperplasia (BPH) is a common disease in elderly men; its occurrence is closely related to the interaction between stromal cells and epithelial cells in the prostate. This article aims to explore the potential therapeutic effect and mechanism of a new trifluoromethyl [...] Read more.
Benign prostatic hyperplasia (BPH) is a common disease in elderly men; its occurrence is closely related to the interaction between stromal cells and epithelial cells in the prostate. This article aims to explore the potential therapeutic effect and mechanism of a new trifluoromethyl quazoline compound (kzl054) on BPH. The results showed that kzl054 had inhibitory activity that limited the growth of prostate hyperplasia cells, BPH-1, and stromal cells, WPMY-1. It could also induce apoptosis of BPH-1 cells and arrest their cell cycle. animal experiment results showed that kzl054 could effectively reduce the volume and prostate index of mouse prostate hyperplasia tissues. Through the establishment of a co-culture system of BPH-1 and WPMY-1 cells, it was found that co-culture could induce EMT in BPH-1 cells. While kzl054 could affect the secretion of TGF-β1 by competitively binding to the colchicine binding site on β-tubulin and inhibiting the expression of β-tubulin, through inhibiting the secretion of TGF-β1 by stromal cells. This study has revealed that compound kzl054 inhibits the secretion of TGF-β1 by targeting the inhibition of microtubule polymerization and regulating the epithelial cell EMT, providing potential candidate molecules and mechanisms for the development of new drugs for the treatment of BPH. Full article
Show Figures

Graphical abstract

13 pages, 1466 KB  
Article
High CD36 Expression Predicts Aggressive Invasion and Recurrence in Oral Squamous Cell Carcinoma
by Kotaro Sakurai, Kei Tomihara, Manabu Yamazaki, Jun-ichi Tanuma and Shin-ichi Yamada
Int. J. Mol. Sci. 2025, 26(24), 12071; https://doi.org/10.3390/ijms262412071 - 15 Dec 2025
Viewed by 174
Abstract
CD36, a fatty acid scavenger receptor expressed in tumors, is associated with a poor prognosis in several cancers. Our previous research demonstrated the involvement of CD36 in the proliferation and migration of oral squamous cell carcinoma (OSCC) cells. However, the clinical significance of [...] Read more.
CD36, a fatty acid scavenger receptor expressed in tumors, is associated with a poor prognosis in several cancers. Our previous research demonstrated the involvement of CD36 in the proliferation and migration of oral squamous cell carcinoma (OSCC) cells. However, the clinical significance of CD36 expression in OSCC remains unclear. The purpose of this study was to evaluate the association between CD36 expression and the clinicopathological characteristics of OSCC patients. Immunohistochemical expression of CD36 was quantified using the H-score, and its association with clinicopathological characteristics was evaluated in 55 OSCC patients. The mean H-score for membrane-associated CD36 expression was 84.8. CD36 expression was significantly correlated with tumor stage, mode of invasion, differentiation, and recurrence of OSCC cells. Moreover, elevated CD36 expression was significantly correlated with a high rate of relapse. Univariate and multivariate analyses showed that CD36 expression was an independent risk factor for relapse. Moreover, The Cancer Genome Atlas (TCGA) dataset analysis revealed that CD36 expression may coexist with transcriptional activation of β-oxidation-related and epithelial–mesenchymal transition (EMT)-related pathways. These findings suggest that CD36 might serve as a predictive biomarker for OSCC malignancy and recurrence. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

18 pages, 1122 KB  
Review
Why and How Are Infants with Hutchinson–Gilford Progeria Syndrome Born Without Severe Manifestations?
by Mariia A. Erokhina, Ekaterina A. Vorotelyak, Andrey V. Vasiliev and Vepa K. Abdyev
Med. Sci. 2025, 13(4), 321; https://doi.org/10.3390/medsci13040321 - 15 Dec 2025
Viewed by 163
Abstract
Children with Hutchinson–Gilford progeria syndrome (HGPS) are born without height and weight abnormalities, and postnatal development is delayed from two months of age. The pathophysiological manifestations of HGPS can be categorized into the three tissue systems that are primarily affected: bone and cartilage, [...] Read more.
Children with Hutchinson–Gilford progeria syndrome (HGPS) are born without height and weight abnormalities, and postnatal development is delayed from two months of age. The pathophysiological manifestations of HGPS can be categorized into the three tissue systems that are primarily affected: bone and cartilage, the smooth muscular layer of the vasculature, and the dermis layer. To understand the biology of the syndrome’s complications resulting from the inherited dominant mutation of the LMNA gene, HGPS has to be considered in embryogenesis. Since the development of the primarily affected HGPS tissues involves a simultaneous contribution of mesodermal and neural crest cells, we hypothesized that the stochastic and heterogeneous coexistence of mesoderm and neural crest cells might be crucial for the onset and manifestation of HGPS. In addition, the expression of Lamin A and/or progerin during embryonic development tends to accumulate in the cell nucleus, causing the syndrome manifestation. Then, how and why are infants with the LMNA gene mutation born without severe deviations? Migration is a distinguishing property of mesoderm and neural crest cells, so that they are continuously subjected to mechanical stimuli throughout development and require normal lamina function. However, the viscoelastic property and the mechanosensor capability to respond to mechanical stress of the HGPS cell nucleus are disturbed. Despite the presence of progerin in development, we assume that high levels of Lamin B1 in cells determine the delayed onset of HGPS after birth. We also hypothesized that progerin toxicity could be managed and prevented, potentially allowing for rescue by the presence of Lamin B1. Full article
Show Figures

Graphical abstract

23 pages, 1039 KB  
Review
Blood-Based miRNA Panels for Timely Detection of Non-Small-Cell Lung Cancer: From Biomarker Discovery to Clinical Translation
by Yazan Zedan, Maria Yurievna Konoshenko, Olga Evgenievna Bryzgunova, Antonina Aleksandrovna Ilyushchenko, Yaroslava Mikhailovna Danilova, Stanislav Dmitrievich Gorbunkov, Kirill Alekseevich Zykov and Pavel Petrovich Laktionov
Int. J. Mol. Sci. 2025, 26(24), 12035; https://doi.org/10.3390/ijms262412035 - 14 Dec 2025
Viewed by 229
Abstract
Lung cancer (LC) remains a leading cause of global cancer mortality, driving the need for novel timely detection strategies, i.e., stages I–II detection when tumor curation is efficient. Circulating microRNA (miRNAs), with their unique stability in biofluids, offer a powerful approach for non-invasive [...] Read more.
Lung cancer (LC) remains a leading cause of global cancer mortality, driving the need for novel timely detection strategies, i.e., stages I–II detection when tumor curation is efficient. Circulating microRNA (miRNAs), with their unique stability in biofluids, offer a powerful approach for non-invasive detection. This review compiles validated miRNAs implicated in the early stages of non-small-cell lung cancer (NSCLC), elucidating their roles in key oncogenic pathways such as epithelial-mesenchymal transition (EMT), PI3K/AKT/mTOR, and JAK-STAT, which regulate proliferation, apoptosis, and metastasis. Furthermore, we critically evaluate developed miRNA panels with a specific focus on advanced quantification and normalization strategies, including exogenous spike-in controls and data-driven methods like pairwise normalization, to enhance diagnostic accuracy. Consequently, we identify and rank the most viable miRNA candidates according to key analytical and clinical metrics, providing a clear roadmap for translating these biomarkers into effective panels for the timely detection of NSCLC. Full article
Show Figures

Figure 1

17 pages, 1461 KB  
Article
Cucurbitacin D Induces Apoptotic Cell Death via NOX4 and Overcomes Radioresistance in Colorectal Cancer
by Tae Woo Kim
Int. J. Mol. Sci. 2025, 26(24), 12022; https://doi.org/10.3390/ijms262412022 - 13 Dec 2025
Viewed by 179
Abstract
Colorectal cancer (CRC) incidence is a significant cancer globally, and radiotherapy resistance is a serious problem. Cucurbitacin D (CBD), extracted from many plants such as the tubers of Trichosanthes kirilowii and the fruits of Ecballium elaterium (squirting cucumber), has various therapeutic effects, such [...] Read more.
Colorectal cancer (CRC) incidence is a significant cancer globally, and radiotherapy resistance is a serious problem. Cucurbitacin D (CBD), extracted from many plants such as the tubers of Trichosanthes kirilowii and the fruits of Ecballium elaterium (squirting cucumber), has various therapeutic effects, such as anti-cancer, -inflammation, -diabetes, and -viral infection effects. Since reports have indicated that CBD exhibits effective anti-cancer activity across various cancer types, our hypothesis is that CBD will overcome radioresistance in CRC radiotherapy. In the present study, we identified that CBD, a triterpenoid compound isolated from Trichosanthes kirilowii and Ecballium elaterium, has an anti-cancer and anti-inflammatory effect in vivo and in vitro. In LPS-induced murine models, CBD suppresses LPS-mediated cytokines, including TNFα, IL-6, IL-1β, and COX-2. In CRC xenograft mouse models, CBD treatment results in significantly smaller tumor volumes than the control. In HCT116 and HT29 cells, CBD treatment suppresses cell viability and increases LDH cytotoxicity and caspase-3 activity and cleavage. However, combined treatment of CBD and Z-VAD-FMK inhibits caspase-dependent apoptosis and cell death. Since CBD induces intracellular calcium (Ca2+) and reactive oxygen species (ROS) generation, it mediates ER stress-induced apoptotic cell death through the PERK-ATF4-CHOP axis. Moreover, ER stress inducer thapsigargin (TG) mediates synergistic apoptotic cell death in CBD-treated HCT116 and HT29 cells. However, PERK or CHOP knockdown suppresses ER stress-mediated apoptosis in CBD-treated HCT116 and HT29 cells. CBD treatment induces oxidative stress through the NADPH Oxidase 4 (NOX4) and also increases ROS generation. However, NOX4 knockdown and ROS inhibitor NAC or DPI block ER stress-induced apoptotic cell death by inhibiting the suppression of cell viability and the elevation of caspase-3 activity, LDH cytotoxicity, and intracellular ROS activity in CBD-mediated HCT116 and HT29 cells. We established radioresistant CRC models (HCT116R and HT29R); subsequently, radiation (2 Gy) in combination with CBD treatment overcame radioresistance via the modulation of the epithelial–mesenchymal transition (EMT) phenomenon, including the increase in N-cadherin and vimentin and the reduction in E-cadherin. Thus, these results show that CBD may be a new powerful therapeutic approach for CRC radiotherapy. Full article
(This article belongs to the Special Issue New Advances in Reversing Cancer Therapy Resistance)
Show Figures

Figure 1

15 pages, 2575 KB  
Article
The Therapeutic Effect of a Biodegradable Long-Acting Intravitreal Implant Containing CGK012 on Neovascular Age-Related Macular Degeneration by Promoting β-Catenin Degradation
by Seoyoung Park, Jihyun Won, Jong Beom Heo, Juhyung Kang, Ye Woon Oh, Geunji Park, Giseong Lee, Jee-Hyun Lee, Gyu-Yong Song, Wonku Kang and Sangtaek Oh
Pharmaceuticals 2025, 18(12), 1884; https://doi.org/10.3390/ph18121884 - 12 Dec 2025
Viewed by 247
Abstract
Background/Objectives: Neovascular age-related macular degeneration (nAMD) poses a serious threat to the eyesight of older adults, representing a leading cause of irreversible vision loss. Anti-vascular endothelial growth factor (anti-VEGF) treatments are effective but require repeated intraocular injections and show poor responses in some [...] Read more.
Background/Objectives: Neovascular age-related macular degeneration (nAMD) poses a serious threat to the eyesight of older adults, representing a leading cause of irreversible vision loss. Anti-vascular endothelial growth factor (anti-VEGF) treatments are effective but require repeated intraocular injections and show poor responses in some patients. CGK012 is a novel derivative of decursin that inhibits the Wnt/β-catenin pathway. This study aimed to elucidate the mode of action of CGK012 and examine its therapeutic effects. Methods: We performed in vitro cellular studies in a retinal pigment epithelial (RPE) cell line (ARPE-19) and human umbilical vein endothelial cells (HUVECs). We examined the in vivo efficacy of CGK012-loaded implants in laser-induced choroidal neovascularization (CNV) rabbit models. We also determined the implants’ in vitro dissolution, intraocular release, and disposition characteristics. Results: CGK012 decreased angiogenic/proinflammatory factor expression and suppressed the epithelial–mesenchymal transition (EMT) in RPE cells by promoting intracellular β-catenin degradation. Additionally, it repressed the expression of cyclin D1 and c-myc, downstream target genes of β-catenin, and inhibited HUVEC capillary tube formation. CGK012-loaded poly (lactic-co-glycolic acid) (PLGA) intravitreal implants significantly reduced vascular leakage in a laser-induced CNV rabbit model. Notably, CGK012 released from the implant was highly permeable to retina/choroid tissue and downregulated β-catenin, angiogenic/inflammatory factors, and vimentin in the rabbit model. The CGK012 concentration reached a plateau at 28–42 days in the vitreous humor and decayed with a half-life of 14 days without systemic exposure. Conclusions: Our findings demonstrate that CGK012 implants prevent choroidal neovascularization through the Wnt/β-catenin pathway suppression and produce high concentrations of CGK012 in the posterior eye segment with prolonged release. Thus, these implants provide more therapeutic choices for nAMD treatment. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

18 pages, 751 KB  
Review
Cancer-Associated Fibroblasts-Derived Exosomes as Mediators of Immunotherapy Resistance in Head and Neck Squamous Cell Carcinoma
by Julia Federspiel, Jozsef Dudas, Benedikt Gabriel Hofauer, Barbara Wollenberg and Teresa Bernadette Steinbichler
Cells 2025, 14(24), 1978; https://doi.org/10.3390/cells14241978 - 12 Dec 2025
Viewed by 257
Abstract
The tumor microenvironment (TME) orchestrates tumor growth, immune evasion, and therapeutic response in head and neck squamous cell carcinoma (HNSCC). Current immune checkpoint inhibitors (ICIs) target the programmed death receptor-1/programmed death-ligand 1 (PD-1/PD-L1) axis and improve survival in recurrent, metastatic, and locally advanced [...] Read more.
The tumor microenvironment (TME) orchestrates tumor growth, immune evasion, and therapeutic response in head and neck squamous cell carcinoma (HNSCC). Current immune checkpoint inhibitors (ICIs) target the programmed death receptor-1/programmed death-ligand 1 (PD-1/PD-L1) axis and improve survival in recurrent, metastatic, and locally advanced HNSCC. Tumor cells produced exosomes directly suppress cytotoxic T-lymphocytes activity by modulating immune checkpoint pathways and disrupting T-cell receptor signaling. Cancer-associated fibroblast-derived exosomes (CAF-Exos) function indirectly by conditioning immune escape and tumor growth. Together, these exosomal populations cooperate to create an immunosuppressive niche that hinders the efficacy of immunotherapies. CAF-Exos induce TME changes that exclude CD8+ T-cells, promote regulatory T-cells (Tregs), and upregulate PD-L1 expression in tumor cells. The bidirectional transfer of microRNAs (miRNAs) between tumor cells and CAFs enhances epithelial–mesenchymal transition (EMT), suppresses cytotoxic lymphocytes, and undermines ICI efficacy. This review article summarizes recent publications about plasma-derived exosomes from HNSCC patients. These exosomes carry tumor and immune checkpoint markers, reflect tumor burden and treatment response, and strongly modulate immune cells by suppressing T- and B-cell activity and promoting immunosuppressive macrophages. We encourage functional and biomechanistic future studies in the field of HNSCC that examine how CAF subtypes exosomes achieve an immunoresistant TME. Full article
Show Figures

Figure 1

Back to TopTop