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 (352)

Search Parameters:
Keywords = cell adhesion and motility

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 4991 KB  
Article
Molecular Basis of Simalikalactone D Sensitivity in Triple-Negative Breast Cancer Cells
by Annelis O. Sánchez-Álvarez, Joshua Nieves-Reyes, Gabriel Borges-Vélez, Josué Pérez-Santiago, Misael Rivera-García, Stella Alicea-Ayala, Claudia Ospina-Millan, Fatima Valiyeva and Pablo E. Vivas-Mejia
Biomolecules 2025, 15(11), 1561; https://doi.org/10.3390/biom15111561 - 6 Nov 2025
Viewed by 388
Abstract
Background/Objective: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) lacking targeted therapies and characterized by high tumor heterogeneity. In this study, we evaluated the anticancer activity and mechanistic profile of Simalikalactone D (SKD), a quassinoid compound derived from the [...] Read more.
Background/Objective: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) lacking targeted therapies and characterized by high tumor heterogeneity. In this study, we evaluated the anticancer activity and mechanistic profile of Simalikalactone D (SKD), a quassinoid compound derived from the endemic Puerto Rican tree Simarouba tulae, in three TNBC cell lines, MDA-MB-468, MDA-MB-231, and SUM-149. Methods: MDA-MB-468, MDA-MB-231 and SUM-149 TNBC cells were evaluated for cell viability, proliferation and migration following SKD treatment. Phospho-antibody array, proteomics, and Western blot analyses were used to explore the SKD mechanism of action in MDA-MB-468 and MDA-MB-231 cell lines. Molecular docking was performed to assess SKD’s interaction with potential intracellular targets. Results: SKD exerted a concentration-dependent effect on the three cell lines. However, MDA-MB-468 cells exhibited an IC50 of 67 nM, while the IC50 values for MDA-MB-231 and SUM-149 were 422 nM and 598 nM, respectively. In MDA-MB-468 cells, 100 nM of SKD induced apoptosis, evidenced by the activated caspase-3 activity, PARP-1 cleavage and decrease in Bcl-2 and survivin protein levels. Sublethal SKD (25 nM) impaired migration in MDA-MB-231 cells and reduced proliferation and motility in SUM149 cells. A 6 h SKD treatment markedly reduced phosphorylation of apoptosis-related proteins (p53, BAD, DAXX, AKT1, JUN) and Jak/STAT pathway components, indicating early disruption of intracellular signaling prior to phenotypic changes. Proteomic profiling showed distinct pathway alterations in both MDA-MB-468 and MDA-MB-231 cells, with reduced Integrin β1 (ITGB1) levels emerging as a shared effector. This suggests that SKD broadly disrupts cell adhesion and migration independently of apoptosis-driven cell death. Western blot validation confirmed reduced ITGB1 protein levels across all three TNBC cell lines examined. In silico docking confirmed favorable binding affinities of SKD to both EGFR (ΔG = −6.718 kcal/mol) and STAT4 (ΔG = −8.481 kcal/mol). Conclusions: Overall, our findings suggest that SKD is a potent anticancer agent in a subgroup of TNBC cells. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

23 pages, 14500 KB  
Article
TFAM Loss Induces Oxidative Stress and Divergent Phenotypes in Glioblastoma Metabolic Subtypes
by Stella G. Cavalcante, Roseli da S. Soares, Miyuki Uno, Maria J. F. Alves, Ricardo C. Cintra, Paula R. Sola, Christiane Y. Ozaki, Antonio M. Lerário, Sueli M. Oba-Shinjo and Suely K. N. Marie
Int. J. Mol. Sci. 2025, 26(21), 10446; https://doi.org/10.3390/ijms262110446 - 27 Oct 2025
Viewed by 455
Abstract
Mitochondrial transcription factor A (TFAM) is essential for mitochondrial DNA (mtDNA) maintenance and function, but its role in glioblastoma (GBM) remains largely unexplored. Analysis of patient astrocytomas and TCGA datasets has revealed progressive TFAM downregulation with increasing malignancy, with the lowest expression in [...] Read more.
Mitochondrial transcription factor A (TFAM) is essential for mitochondrial DNA (mtDNA) maintenance and function, but its role in glioblastoma (GBM) remains largely unexplored. Analysis of patient astrocytomas and TCGA datasets has revealed progressive TFAM downregulation with increasing malignancy, with the lowest expression in glycolytic/plurimetabolic (GPM) subtypes. Functional and transcriptomic profiling of mesenchymal GBM cell lines showed that TFAM silencing in GPM-type U87MG cells enhanced proliferation, S-phase entry, reactive oxygen species (ROS) production, and adhesion, while reducing motility. These changes were correlated with upregulation of LDHC and TRAF2 and downregulation of androgen receptor-linked motility genes and LOXL2. By contrast, TFAM loss in mitochondrial (MTC)-type A172 cells caused minimal phenotypic alterations, associated with elevated SOD1 expression and activation of antioxidant, mitochondrial membrane, and survival pathways, alongside suppression of oxidative phosphorylation and vesicle-trafficking genes. TFAM overexpression reduced proliferation in U87MG but had a limited impact on A172 cells. Taken together, these findings establish TFAM as a subtype-specific regulator of GBM cell proliferation, redox balance, and motility. TFAM loss drives a proliferative, ROS-sensitive phenotype in GPM-type cells, while eliciting adaptive, stress-resilient programs in MTC-type cells. This study identifies TFAM and downstream effectors, TRAF2 and LOXL2, as potential therapeutic targets, supporting the development of metabolic subtype-tailored strategies for GBM treatment. Full article
(This article belongs to the Special Issue New Players in the Research of Oxidative Stress and Cancer)
Show Figures

Figure 1

13 pages, 2925 KB  
Article
Volatile Compounds as Upcoming Antigiardial Agents: In Vitro Action of Carvacrol, Thymol and p-Cymene on Giardia lamblia Trophozoites
by Marisa Machado, Ana Silva, Rui Linhares, Carlos Cavaleiro and Maria C. Sousa
Pharmaceutics 2025, 17(11), 1380; https://doi.org/10.3390/pharmaceutics17111380 - 24 Oct 2025
Viewed by 467
Abstract
Background/Objectives: Carvacrol and thymol are monoterpenes present in phenolic-rich essential oils extracted from aromatic plants that exhibit antimicrobial activity. This study evaluates the antiprotozoal effect of carvacrol, thymol and their precursor, p-Cymene, against Giardia lamblia and investigates their mechanism of action and cytotoxicity [...] Read more.
Background/Objectives: Carvacrol and thymol are monoterpenes present in phenolic-rich essential oils extracted from aromatic plants that exhibit antimicrobial activity. This study evaluates the antiprotozoal effect of carvacrol, thymol and their precursor, p-Cymene, against Giardia lamblia and investigates their mechanism of action and cytotoxicity profile. Methods: G. lamblia susceptibility, cell viability, swelling and adhesion abilities following application of carvacrol, thymol and p-Cymene were assessed. Ultrastructural changes were evaluated using electron microscopy. Cytotoxicity was determined in mammalian cell lines (murine macrophages RAW 264.7 and bovine aortic endothelial cells) exposed to the same IC50 concentrations effective against G. lamblia. Results: Carvacrol and thymol led to significant inhibition of G. lamblia trophozoite proliferation (IC50 ≅ 50 µg/mL). After 7 h of incubation, total cell number decreased by 30% (p < 0.01) with carvacrol and by 50% (p < 0.001) with thymol, accompanied by reduced motility and adhesion (<20% attached cells). At IC50 concentrations, G. lamblia trophozoites exposed to carvacrol and thymol underwent considerable ultrastructural alterations (e.g., aberrant-shaped cells, mitochondrial swelling and autophagosomal structures). Reduced trophozoite motility and adhesion capacity were also observed. In mammalian cells, thymol showed no significant cytotoxicity, whereas carvacrol significantly reduced viability in both cell lines. In contrast, p-Cymene showed no antigiardial activity. Conclusions: Our data suggests that carvacrol and thymol disrupt G. lamblia trophozoite integrity, possibly through alterations in membrane permeability and osmoregulatory processes. In conclusion, these compounds reveal in vitro antigiardial activity, supporting their potential as antigiardial drugs. Full article
Show Figures

Figure 1

31 pages, 3420 KB  
Systematic Review
From Adhesion to Invasion: Integrins, Focal Adhesion Signaling, and Actin Binding Proteins in Cervical Cancer Progression—A Scoping Review
by Marta Hałas-Wiśniewska, Patryk Zawadka, Wioletta Arendt and Magdalena Izdebska
Cells 2025, 14(20), 1640; https://doi.org/10.3390/cells14201640 - 21 Oct 2025
Viewed by 815
Abstract
Background: Cervical cancer (CC) is one of the most common malignancies in women worldwide. Its progression involves a cascade of processes, including proliferation, migration, invasion, and metastasis. Each stage is regulated by specific signaling pathways. Objective: This scoping review aimed to map current [...] Read more.
Background: Cervical cancer (CC) is one of the most common malignancies in women worldwide. Its progression involves a cascade of processes, including proliferation, migration, invasion, and metastasis. Each stage is regulated by specific signaling pathways. Objective: This scoping review aimed to map current evidence on the role of cell adhesion-related molecules, including integrins, focal adhesion (FA) proteins, and actin-binding proteins (ABPs), in CC progression. These protein groups act in a coordinated manner—integrins perceive and transmit extracellular matrix (ECM) signals, FA proteins mediate intracellular signaling, and ABPs reorganize the cytoskeleton, ensuring the continuity of adhesion and motility processes. Methods: A structured literature search was conducted for studies published between 2015 and 2025. Eligible articles described the role of adhesion-related proteins in migration, invasion, or EMT in CC. Data were synthesized thematically according to protein families. Results: The evidence highlights integrins, FA/FAK, and ABPs as interconnected regulators coordinating ECM signaling and cytoskeletal remodeling during CC progression. Their dysregulation is associated with enhanced migration, EMT induction, angiogenesis, and therapy resistance. Conclusions: This review provides a unique, integrated perspective linking adhesion molecules with invasion mechanisms in CC progression, providing new insights into their interplay. Understanding the interaction between these proteins is therefore a crucial step in the treatment of CC and may facilitate the discovery of biomarkers and support the development of targeted therapies. Full article
Show Figures

Figure 1

24 pages, 1454 KB  
Review
The Role of Tenascin-C in Neuroinflammation and Neuroplasticity
by Ya-Li Jin, Shi-Wen Bao, Meng-Xuan Huang, Yong-Jing Gao, Huan-Jun Lu and Xiao-Bo Wu
Int. J. Mol. Sci. 2025, 26(20), 10174; https://doi.org/10.3390/ijms262010174 - 19 Oct 2025
Viewed by 730
Abstract
Tenascin-C (TNC) is a complex extracellular matrix (ECM) protein that plays a critical role in regulating cellular adhesion, motility, proliferation, and inflammation through its interaction with Toll-like receptor 4 (TLR4) and other receptors. The upregulation of TNC is associated with inflammatory responses, autoimmune [...] Read more.
Tenascin-C (TNC) is a complex extracellular matrix (ECM) protein that plays a critical role in regulating cellular adhesion, motility, proliferation, and inflammation through its interaction with Toll-like receptor 4 (TLR4) and other receptors. The upregulation of TNC is associated with inflammatory responses, autoimmune disorders, and neoplastic conditions during both physiological and pathological tissue remodeling. In the central nervous system (CNS), TNC contributes to neuroinflammatory processes by modulating the function of immune cells and the secretion of pro-inflammatory mediators, thereby playing a pivotal role in the initiation and progression of neuroinflammatory diseases. TNC is expressed in astrocytes, neural progenitor cells, and various neuronal populations within both developing and mature CNS regions. It regulates neuronal migration and axonal guidance during neurogenesis, facilitating synaptic plasticity and CNS regeneration. Furthermore, TNC enhances neuroplasticity through interactions with receptor families, such as integrins, to establish the molecular connections necessary for cell communication and signal transduction. This review investigates the mechanistic properties of TNC, focusing on its spatiotemporal expression, molecular interactions with receptors, and its role in neurological disorders, in addition to its modulatory capacity in neuroplastic processes. Additionally, this review delves into recent research advancements with respect to neuroinflammation involving TNC, along with therapeutic strategies targeting TNC. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanism in Neuroinflammation Research)
Show Figures

Figure 1

17 pages, 1489 KB  
Article
Embryonic Benzo[a]pyrene Exposure Induces Multigenerational Reproductive Effects on Adult Male Medaka: Phenotypic and Transcriptomic Insights
by Yinhua Chen, Yi Yang, Xian Qin, Jiangang Wang, Guanglong Tang, Rim EL Amouri, Jiayang Chen, Jack Chi-Ho Ip, Wenhua Liu and Jiezhang Mo
Toxics 2025, 13(10), 886; https://doi.org/10.3390/toxics13100886 - 17 Oct 2025
Viewed by 429
Abstract
Benzo[a]pyrene (B[a]P), a ubiquitous environmental pollutant, poses a significant threat to male reproductive health, but the underlying latent molecular mechanisms remain virtually unknown. This study investigated the effects of embryonic B[a]P exposure on testicular function and spermatogenesis in F0 and F1 adult male [...] Read more.
Benzo[a]pyrene (B[a]P), a ubiquitous environmental pollutant, poses a significant threat to male reproductive health, but the underlying latent molecular mechanisms remain virtually unknown. This study investigated the effects of embryonic B[a]P exposure on testicular function and spermatogenesis in F0 and F1 adult male medaka (Oryzias latipes). Embryos were exposed to sublethal concentrations (2.5, 20, and 80 μg/L) for 8 days and then raised in clean water until they reached adulthood. Transcriptomic analysis of F0 testicular tissues revealed widespread dysregulation of critical pathways. Exposure impaired the brain–pituitary–gonadal axis by disrupting GnRH signaling and downregulating genes encoding key steroidogenic enzymes (CYP17A1, HSD3B2), indicating suppressed testosterone biosynthesis. Concurrently, pathways essential for cellular energy metabolism (AMPK signaling, insulin signaling), amino acid biosynthesis, and cytoskeletal organization (actin cytoskeleton, focal adhesion) were profoundly altered. Furthermore, B[a]P activated apoptotic pathways and disrupted the balance between cell survival (PI3K-Akt signaling) and death, compromising spermatogenic cell fate. These molecular disruptions manifested in drastic physiological impairments, including a reduced gonadosomatic index, decreased sperm motility, and compromised fertilization success in F0 males, although these effects were recovered in the F1 generation. This study provides a comprehensive molecular basis for the long-term reproductive toxicity of early-life B[a]P exposure. Full article
(This article belongs to the Special Issue Environmental Contaminants and Human Health—2nd Edition)
Show Figures

Figure 1

14 pages, 4433 KB  
Article
Saucerneol D Suppresses the Growth of Helicobacter pylori and Their Virulence Factors
by Su Man Kim, Hyun Jun Woo, Zhongduo Yang, Tiankun Zhao, Ji Yeong Yang and Sa-Hyun Kim
Curr. Issues Mol. Biol. 2025, 47(10), 828; https://doi.org/10.3390/cimb47100828 - 9 Oct 2025
Viewed by 402
Abstract
Helicobacter pylori infects the human stomach and causes various gastrointestinal diseases. Saucerneol D is a type of lignan, which is a polyphenol compound that exists naturally in plants, and it is abundant in flaxseed, sesame seeds, whole grains, vegetables, and fruits. Saucerneol D [...] Read more.
Helicobacter pylori infects the human stomach and causes various gastrointestinal diseases. Saucerneol D is a type of lignan, which is a polyphenol compound that exists naturally in plants, and it is abundant in flaxseed, sesame seeds, whole grains, vegetables, and fruits. Saucerneol D is found in Saurus chinensis extract and has been reported to exert a variety of effects, such as antioxidant and anti-inflammatory abilities. However, its antibacterial effect against H. pylori has not been reported; therefore, we analyzed the effect of saucerneol D on H. pylori in the present study. Changes in the expression of pathogenic factors and gene transcription in H. pylori were observed after treatment with saucerneol D using Western blotting and RT-PCR. It was confirmed that saucerneol D suppressed the growth of H. pylori by decreasing the expression of the genes dnaN and polA, which are required for bacterial replication. Saucerneol D also reduced the secretion of the major pathogenic toxin protein, CagA, by downregulating the expression of type IV secretion system-composing proteins. Furthermore, saucerneol D reduced ammonia production by inhibiting the expression of urease proteins, which are essential for the survival of H. pylori in the acidic gastric environment. Additionally, saucerneol D decreased the expression of flaB, potentially reducing motility. Finally, it was confirmed that the expression of the sabA gene, associated with cell adhesion, was reduced. These results suggest that saucerneol D inhibits the growth of H. pylori and the expression of several pathogenic factors, indicating that saucerneol D has an antimicrobial effect against H. pylori. Full article
Show Figures

Graphical abstract

16 pages, 2654 KB  
Article
Differential Sensitivity to MEK Inhibitors Highlights Distinct Entosis Mechanisms in BxPC3 and MCF7 Cells
by Paweł Tyrna, Julia Kostro, Monika Olszanecka, Piotr Szukało and Izabela Młynarczuk-Biały
Cells 2025, 14(19), 1500; https://doi.org/10.3390/cells14191500 - 25 Sep 2025
Viewed by 508
Abstract
Entosis is a form of cell-in-cell interaction observed in epithelial cancers, characterized by the internalization of one cell into another. This process is initiated by cell detachment, cadherin-mediated homotypic adhesion, and the formation of an entotic vacuole. Mechanistically, entosis is driven by Rho/ROCK [...] Read more.
Entosis is a form of cell-in-cell interaction observed in epithelial cancers, characterized by the internalization of one cell into another. This process is initiated by cell detachment, cadherin-mediated homotypic adhesion, and the formation of an entotic vacuole. Mechanistically, entosis is driven by Rho/ROCK signaling and actomyosin contractility in the invading (inner) cell, which becomes stiffer and is pulled into the softer host (outer) cell. A functional assay using differently stained cell populations allows for the assessment of pharmacological interventions on either the inner or outer cell during entosis. In this study, we investigated the impact of MEK pathway inhibition on entosis in two epithelial cancer cell lines, BxPC3 (pancreatic cancer) and MCF7 (breast cancer). BxPC3 cells, which rely on adhesion, exhibited a significant reduction in entotic index upon MEK inhibition. In contrast, MCF7 cells showed no selectivity of entosis to three different MEK inhibitors. These findings suggest cell-type-specific regulation of entosis, potentially linked to differences in protrusion formation mechanisms and upstream Ras signaling pathways previously implicated in cancer cell motility. Full article
(This article belongs to the Topic Kinases in Cancer and Other Diseases, 2nd Edition)
Show Figures

Figure 1

21 pages, 6074 KB  
Article
Therapeutic Potential of Probiotic-Derived P8 Protein as an Anti-Metastatic Agent in Colorectal Cancer
by Byung Chull An, Seungwoo Kim, Jaewon Ha, Sang-Hyuk Seok, Jun Won Park, Yongku Ryu and Myung Jun Chung
Microorganisms 2025, 13(9), 2175; https://doi.org/10.3390/microorganisms13092175 - 17 Sep 2025
Viewed by 596
Abstract
We previously described the use of probiotics to deliver a Lactobacillus rhamnosus-derived therapeutic protein, P8, which has been identified as a candidate colorectal cancer (CRC) suppressor protein with anti-proliferation and anti-migration activities. P8 was found to penetrate cell membranes by endocytosis, suppressing [...] Read more.
We previously described the use of probiotics to deliver a Lactobacillus rhamnosus-derived therapeutic protein, P8, which has been identified as a candidate colorectal cancer (CRC) suppressor protein with anti-proliferation and anti-migration activities. P8 was found to penetrate cell membranes by endocytosis, suppressing cell proliferation through G2 cell cycle arrest. Despite the ability of P8 to suppress cell migration in vitro, its mechanism of action in CRC is unclear. We profiled the P8-interacting partner proteins using the pull-down method with His-tagged bait P8 and then identified them by LC-MS/MS. Among the interacting targets, we focused on the mothers against decapentaplegic homolog 1 (Smad1), which is well known as one of the important modulators of the bone morphogenetic protein (BMP)-derived migration pathway in CRC. The present study discovers that P8 prevents the phosphorylation of Smad1 or heterologous complexes within the Smad family, interfering with the importation of Smad1 or its complexes into the nucleus. Thus, P8 significantly inhibits the up-regulation of epithelial–mesenchymal transition (EMT)-related genes mediated by Smad1. P8 also inhibits the morphological changes required for cell migration or adhesion. P8 induces morphologic changes in DLD-1 cells, and their spheroid surfaces, resulting in a significant reduction of the number and length of filopodia, as well as the down-regulation of the expression of myosin X and its accumulation in filopodia tips. This phenomenon seems to be a major negative regulator of cell motility that could be of key importance in metastasis. Use of a mouse model of human CRC metastasis confirmed that P8 significantly suppresses the liver metastatic rate. Probiotic-derived protein P8 significantly suppresses CRC metastasis through inhibition of the Smad1-EMT signal pathway and cell–cell adhesion. Full article
(This article belongs to the Topic News and Updates on Probiotics)
Show Figures

Figure 1

15 pages, 1316 KB  
Review
The Role of Pyk2 Kinase in Glioblastoma Progression and Therapeutic Targeting
by Lilia Kucheryavykh and Yuriy Kucheryavykh
Cancers 2025, 17(16), 2611; https://doi.org/10.3390/cancers17162611 - 9 Aug 2025
Viewed by 845
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor with limited treatment options and poor prognosis. Proline-rich tyrosine kinase 2 (Pyk2) has been implicated in regulation of GBM invasion, proliferation, and recurrence. Its activation, driven by tumor-infiltrating microglia and macrophage-derived extracellular factors such as [...] Read more.
Glioblastoma (GBM) is a highly aggressive brain tumor with limited treatment options and poor prognosis. Proline-rich tyrosine kinase 2 (Pyk2) has been implicated in regulation of GBM invasion, proliferation, and recurrence. Its activation, driven by tumor-infiltrating microglia and macrophage-derived extracellular factors such as EGF, PDGFB, SDF-1α, IL-6, and IL-8, enhances tumor cell motility and survival. Experimental studies demonstrate that pharmacological inhibition or genetic knockdown of Pyk2 significantly reduces glioma cell migration and proliferation. Furthermore, recurrent GBM tumors exhibit elevated Pyk2 phosphorylation in mouse GBM models, correlating with increased tumor growth. Inhibition of Pyk2 and the structurally related focal adhesion kinase (FAK) signaling has shown promising results in preclinical studies, reducing tumor recurrence and improving survival outcomes. This review summarizes recent findings and underscores the pivotal role of Pyk2 in GBM pathophysiology, highlighting its potential as a therapeutic target. Full article
Show Figures

Figure 1

23 pages, 26085 KB  
Article
How Actin Polymerization and Myosin II Activity Regulate Focal Adhesion Dynamics in Motile Cells
by Anastasiia Kovaleva, Evgeniya Solomatina, Madina Tlegenova, Aleena Saidova and Ivan A. Vorobjev
Int. J. Mol. Sci. 2025, 26(16), 7701; https://doi.org/10.3390/ijms26167701 - 9 Aug 2025
Viewed by 1531
Abstract
Focal adhesions (FAs) are multi-protein complexes that mediate cell attachment to the extracellular matrix. Their formation and maturation depend on intracellular tension generated by actin filaments interacting with phosphorylated myosin II. Using live-cell and confocal microscopy, we investigated how FA dynamics are regulated [...] Read more.
Focal adhesions (FAs) are multi-protein complexes that mediate cell attachment to the extracellular matrix. Their formation and maturation depend on intracellular tension generated by actin filaments interacting with phosphorylated myosin II. Using live-cell and confocal microscopy, we investigated how FA dynamics are regulated by actin polymerization and myosin II-driven contractility. We found that knockdown of myosin II resulted in complete and irreversible disassembly of FAs. However, partial inhibition of myosin II, through either ROCK or myosin light chain kinase (MLCK) inhibitors, leads to gradual FA shrinkage. In contrast, complete inhibition of myosin II phosphorylation causes disassembly of existing FAs, followed by the formation of new, small FAs at the cell periphery. In both cases, FAs formed after inhibition of myosin II phosphorylation exhibited significantly longer lifespans than FAs in control cells. Similarly, partial inhibition of actin polymerization using nanomolar concentrations of latrunculin B or cytochalasin D also promoted the formation of small FAs. Complete and irreversible FA disassembly occurred only when actin filaments were fully disrupted, leading to cell lamella retraction. These findings suggest that actin polymerization at the cell edge is the minimal and sufficient requirement for the assembly of small FAs. Notably, our data demonstrate for the first time that perturbation of the actin–myosin system results in stabilization and prolonged lifespan of small FAs, whereas larger FAs, formed in the presence of myosin II activity, are more dynamic. Together, these results emphasize the essential role of cortical actin organization and myosin II phosphorylation in the maintenance and turnover of FAs. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

14 pages, 5071 KB  
Article
Bioactive Phenolics from Vinegar–Egg Accelerates Acute Wound Healing by Activation of Focal Adhesion and Mitogen-Activated Protein Kinase Signaling
by Taehoon Oh, Chan Hee Cho, Su Cheol Baek, Mun Seok Jo, Woo Bong Kang, Yun Seok Kang, Sung-Kyun Ko and Ki Hyun Kim
Nutrients 2025, 17(16), 2584; https://doi.org/10.3390/nu17162584 - 8 Aug 2025
Viewed by 856
Abstract
Background/Objectives: Vinegar–egg is a traditional health-promoting beverage prepared by soaking eggs in vinegar. While both eggs and vinegar are common dietary components with well-documented nutritional and pharmacological activities, eggs treated with vinegar have been rarely studied. This study aims to identify and characterize [...] Read more.
Background/Objectives: Vinegar–egg is a traditional health-promoting beverage prepared by soaking eggs in vinegar. While both eggs and vinegar are common dietary components with well-documented nutritional and pharmacological activities, eggs treated with vinegar have been rarely studied. This study aims to identify and characterize bioactive compounds in vinegar–egg and investigate their potential wound-healing activities. Methods: The vinegar–egg extract was analyzed using liquid chromatography–mass spectrometry (LC–MS) and column chromatography, including HPLC purification, which led to the isolation of four phenolic compounds. Results: These compounds were identified as 4-hydroxybenzoic acid (1), vanillic acid (2), methyl syringate (3), and leptosperin (4) using ESI-MS, UV, and NMR spectroscopic data. Among the isolates, 4-hydroxybenzoic acid (1) and vanillic acid (2) demonstrated wound-healing properties in mouse embryonic fibroblast (MEF) cells. None of the compounds, 4-hydroxybenzoic acid (1), vanillic acid (2), methyl syringate (3), or leptosperin (4), exhibited cytotoxicity in PC12, AGS, MEF, or MDA-MB-231 cells. Notably, 4-hydroxybenzoic acid (1) enhanced cell motility by 2.59-fold and cell invasion by 1.20-fold, while vanillic acid (2) increased cell motility by 2.69-fold and cell invasion by 1.23-fold. Western blot analysis revealed that treatment with 4-hydroxybenzoic acid (1) and vanillic acid (2) increased the phosphorylation of focal adhesion kinase (p-FAK) and matrix metalloproteinase 2 (MMP-2). Furthermore, both compounds elevated the phosphorylation of p38, a key regulator in wound-healing pathways. Conclusions: These findings demonstrate that 4-hydroxybenzoic acid (1) and vanillic acid (2) accelerate wound healing through the activation of focal adhesion and mitogen-activated protein kinase (MAPK) signaling pathways. These results highlight vinegar–egg as a promising therapeutic candidate for wound healing. Full article
(This article belongs to the Special Issue Effects of Plant Extracts on Human Health—2nd Edition)
Show Figures

Figure 1

17 pages, 6016 KB  
Article
Role of Kindlin-2 in Cutaneous Squamous Carcinoma Cell Migration and Proliferation: Implications for Tumour Progression
by Anamika Dutta, Michele Calder and Lina Dagnino
Int. J. Mol. Sci. 2025, 26(15), 7426; https://doi.org/10.3390/ijms26157426 - 1 Aug 2025
Viewed by 492
Abstract
The Kindlin family of scaffold proteins plays key roles in integrin-mediated processes. Kindlin-1 and -2, encoded by the FERMT1 and FERMT2 genes, respectively, are expressed in the epidermis. Kindlin-1 plays protective roles against the development of cutaneous squamous cell carcinomas (cSCCs) in epidermal [...] Read more.
The Kindlin family of scaffold proteins plays key roles in integrin-mediated processes. Kindlin-1 and -2, encoded by the FERMT1 and FERMT2 genes, respectively, are expressed in the epidermis. Kindlin-1 plays protective roles against the development of cutaneous squamous cell carcinomas (cSCCs) in epidermal keratinocytes. However, the role of Kindlin-2 in transformed epidermal keratinocytes has remained virtually unexplored. In this study, we used siRNA approaches to generate Kindlin-2-depleted cells in three isogenic transformed keratinocyte lines. PM1, MET1, and MET4 cells model, respectively, a precancerous lesion, a primary cSCC, and a metastatic lesion of the latter. MET1 cells express both Kindlin-1 and -2. However, Kindlin-1 was not detectable in PM1 and MET4 cells. FERMT2 silencing in PM1 and MET4, but not in MET1 cells, reduced proliferation and the ability to adhere to culture surfaces and spreading. Furthermore, Kindlin-2-depleted PM1 and MET4, but not MET1 cells, exhibited decreased numbers of focal adhesions, as well as an altered F-actin and microtubule cytoskeletal organization. Significantly, FERMT2 silencing reduced the directional migration in all three cell types. These findings are consistent with the concept that, in the absence of other Kindlin orthologues, Kindlin-2 plays a prominent role in the modulation of the proliferation, spreading, focal adhesion assembly, and motility of transformed keratinocytes, as exemplified by PM1 and MET4 cells. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

25 pages, 3835 KB  
Article
A Marine-Derived Steroid from Rhodococcus sp., 3,12-Dioxochola-4,6-dien-24-oic Acid, Enhances Skin Re-Epithelialization and Tissue Repair
by Mücahit Varlı, Hui Tan, Chaeyoung Lee, Jeongyun Lee, Ji Young Lee, Jeong-Hyeon Kim, Songyi Lee, Hangun Kim and Sang-Jip Nam
Mar. Drugs 2025, 23(7), 292; https://doi.org/10.3390/md23070292 - 19 Jul 2025
Viewed by 1327
Abstract
The discovery of bioactive natural compounds from microbes holds promise for regenerative medicine. In this study, we identified and characterized a steroid-like compound, 3,12-dioxochola-4,6-dien-24-oic acid (DOCDA), from a crude extract of Rhodococcus sp. DOCDA significantly promoted wound healing by enhancing HaCaT cell invasion [...] Read more.
The discovery of bioactive natural compounds from microbes holds promise for regenerative medicine. In this study, we identified and characterized a steroid-like compound, 3,12-dioxochola-4,6-dien-24-oic acid (DOCDA), from a crude extract of Rhodococcus sp. DOCDA significantly promoted wound healing by enhancing HaCaT cell invasion and migration. It upregulated key growth factors (EGF, VEGF-A, IGF, TGF-β, and HGF), indicating the activation of regenerative signaling. Additionally, DOCDA increased the expression of genes related to focal adhesion and cytoskeletal regulation (ITGB1, ITGA4, FAK, SRC, RHOA, CDC42, RAC1, and paxillin), supporting enhanced cellular motility and remodeling. Notably, DOCDA promoted stem-like properties in HaCaT cells, as shown by increased spheroid formation and elevated levels of the stemness markers ALDH1 and CD44. Target prediction and molecular docking identified the glucocorticoid receptor (GR) as the primary target of DOCDA, with a docking score of −7.7 kcal/mol. Network and pathway enrichment analysis revealed that GR-linked pathways were significantly associated with wound healing, including steroid hormone signaling, inflammation, immune responses, and cell migration. In vivo, the topical application of DOCDA led to over 70% wound closure in mice by day 5. These findings suggest that DOCDA is a steroid-like compound that accelerates wound healing and may serve as a potential agent in regenerative therapy. Full article
(This article belongs to the Section Structural Studies on Marine Natural Products)
Show Figures

Graphical abstract

12 pages, 5644 KB  
Article
A Subset of HOX Genes Negatively Correlates with HOX/PBX Inhibitor Target Gene Expression and Is Associated with Apoptosis, DNA Repair, and Metabolism in Prostate Cancer
by Richard Morgan, Christopher Smith and Hardev Pandha
Genes 2025, 16(7), 824; https://doi.org/10.3390/genes16070824 - 15 Jul 2025
Viewed by 835
Abstract
Background/Objectives: The HOX genes encode a family of homeodomain-containing transcription factors that have important roles in defining cell and tissue identity in embryonic development, but which also show deregulated expression in many cancers and have been shown to have pro-oncogenic roles. Due to [...] Read more.
Background/Objectives: The HOX genes encode a family of homeodomain-containing transcription factors that have important roles in defining cell and tissue identity in embryonic development, but which also show deregulated expression in many cancers and have been shown to have pro-oncogenic roles. Due to their functionally redundant nature, strategies to target HOX protein function in cancer have focused on their interaction with their PBX cofactor using competitive peptides such as HXR9. HOX/PBX inhibition triggers apoptosis through a sudden increase in target gene expression, including Fos, DUSP1, and ATF3, which are otherwise repressed by HOX/PBX binding. Methods: We analyzed publicly available transcriptomic data in the R2 platform. Results: We show that a specific subgroup of HOX genes is negatively correlated with Fos, DUSP1, and ATF3 expression in prostate cancer, and that this subgroup also shows a strong positive corelation with pathways that support tumour growth, most notably DNA repair and aminoacyl tRNA biosynthesis, and a negative correlation with genes that promote cell adhesion and prevent motility. In addition, this set of HOX genes strongly correlates with patient age, reflecting a previously identified progressive loss of regulation of HOX expression in normal peripheral blood cells. Conclusions: Our findings indicate these HOX genes may have pro-oncogenic functions in prostate cancer. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Back to TopTop