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
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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,189)

Search Parameters:
Keywords = WNT16

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 5436 KB  
Article
MicroRNAs Let-7b-5p and miR-24-3p as Potential Therapeutic Agents Targeting Pancreatic Cancer Stem Cells
by Maricela Medrano-Silva, Eric Genaro Salmerón-Bárcenas, Elena Arechaga-Ocampo, Nicolas Villegas-Sepúlveda, Leopoldo Santos-Argumedo, Sonia Mayra Pérez-Tapia, Mayte Lizeth Padilla-Cristerna, Georgina Hernández-Montes, Gabriela Hernández-Galicia, Ana Beatriz Sánchez-Argáez, Paola Briseño-Díaz, Carmen Sánchez-Torres, Arturo Aguilar-Rojas, Andrea Martínez-Zayas, Miguel Vargas and Rosaura Hernández-Rivas
Int. J. Mol. Sci. 2025, 26(22), 11066; https://doi.org/10.3390/ijms262211066 (registering DOI) - 15 Nov 2025
Abstract
Pancreatic cancer poses a major clinical challenge due to its aggressiveness, frequent recurrence, and limited response to current chemotherapeutic approaches. Cancer stem cells (CSCs), particularly pancreatic CSCs (PCSCs), are key drivers of tumor initiation, therapeutic resistance, and disease relapse. MicroRNAs (miRNAs) have emerged [...] Read more.
Pancreatic cancer poses a major clinical challenge due to its aggressiveness, frequent recurrence, and limited response to current chemotherapeutic approaches. Cancer stem cells (CSCs), particularly pancreatic CSCs (PCSCs), are key drivers of tumor initiation, therapeutic resistance, and disease relapse. MicroRNAs (miRNAs) have emerged as critical regulators of CSC biology and influence self-renewal, pluripotency, and drug resistance through key signaling pathways. To identify PCSC-specific miRNAs, we enriched these cells using the pancreosphere culture method and isolated PCSC+ and PCSC− populations using FACS based on their expression of CD44, CD24, and CD133 surface markers. MicroRNA microarray analysis revealed 31 differentially expressed miRNAs (DEmiRNAs), of which 10 downregulated miRNAs were involved in pathways regulating pluripotency, including the Wnt/β-catenin, TGF-β, MAPK, and PI3K/AKT pathways. Then, 2 of these 10 DEmiRNAs, let-7b-5p and miR-24-3p, were selected for experimental validation. Their overexpression in PCSC+ cells inhibited these pathways, downregulated pluripotency factors, and induced differentiation into endocrine and exocrine phenotypes, as confirmed by RT-qPCR, Western blot, and RNA-seq. Functionally, each miRNA reduced sphere formation, increased gemcitabine sensitivity, and suppressed tumorigenicity in vivo, highlighting their potential as therapeutic candidates. Restoring tumor-suppressive miRNA expression may offer a novel strategy to overcome chemoresistance and improve outcomes in pancreatic cancer. Full article
(This article belongs to the Special Issue MicroRNAs in Physiology and Pathophysiology)
Show Figures

Figure 1

18 pages, 970 KB  
Article
Divergent Tissue and Circulatory Expression of miR-10a in Canine Hepatocellular Carcinoma: Comparative Insights from Human HCC
by Most Shumi Akhter Shathi, Mohammad Arif, Nobuhiro Nozaki, Yutaro Ide, Yoshiyuki Akiyama, Shaohsu Wang, Masashi Takahashi and Naoki Miura
Curr. Issues Mol. Biol. 2025, 47(11), 950; https://doi.org/10.3390/cimb47110950 (registering DOI) - 15 Nov 2025
Abstract
Canine hepatocellular carcinoma (HCC), the most common primary liver malignancy in dogs, shares many clinicopathological and molecular similarities with human HCC. However, its molecular characteristics remain insufficiently defined, and reliable diagnostic biomarkers are lacking. Elucidating dysregulated microRNAs (miRNAs) may aid in both disease [...] Read more.
Canine hepatocellular carcinoma (HCC), the most common primary liver malignancy in dogs, shares many clinicopathological and molecular similarities with human HCC. However, its molecular characteristics remain insufficiently defined, and reliable diagnostic biomarkers are lacking. Elucidating dysregulated microRNAs (miRNAs) may aid in both disease characterization and comparative oncology research. Small RNA sequencing datasets from canine HCC were analyzed to identify significantly dysregulated miRNAs with high expression and biomarker potential. The top candidate was validated in clinical tissues, cell lines, patient’s plasma and plasma exosomes using RT-qPCR. Comparative analyses were conducted using human HCC datasets (TCGA and GEO), followed by target prediction and functional enrichment to identify conserved molecular pathways. Among the 59 differentially expressed miRNAs, cfa-miR-10a showed the highest average expression level and yet was significantly downregulated in canine HCC tissues. RT-qPCR confirmed reduced expression of cfa-miR-10a in canine HCC tissues, whereas plasma exosomes showed significant enrichment, demonstrating excellent diagnostic performance (AUC = 0.94). The mature sequence of cfa-miR-10a is highly conserved with hsa-miR-10a-5p. TCGA datasets confirmed downregulation of hsa-miR-10a-5p in HCC tissues, whereas a GEO dataset showed no significant change in serum exosome levels. Target prediction and functional annotation identified 59 overlapping genes, with the Proteoglycans in cancer pathways being conserved in both species, mediated by ACTG1, SDC1, FRS2, and WNT9B. Collectively, these findings demonstrate distinct intra-tumoral and exosomal expression pattern of miR-10a in canine HCC and support its potential as a non-invasive biomarker with translational relevance. Full article
26 pages, 6764 KB  
Article
Integrative Transcriptome Analysis Across Follicles Highlights Key Regulatory Pathways in Low and High-Egg-Laying Hens
by Armughan Ahmed Wadood, Farhad Bordbar and Xiquan Zhang
Animals 2025, 15(22), 3300; https://doi.org/10.3390/ani15223300 (registering DOI) - 15 Nov 2025
Abstract
Egg-laying performance in hens is regulated by complex molecular mechanisms within the hypothalamic–pituitary–gonadal (HPG) axis and ovarian follicles. This study employed integrative transcriptome profiling of primordial (PR), primary (PM), small white (SW), and small yellow (SY) follicles in hens with low and high [...] Read more.
Egg-laying performance in hens is regulated by complex molecular mechanisms within the hypothalamic–pituitary–gonadal (HPG) axis and ovarian follicles. This study employed integrative transcriptome profiling of primordial (PR), primary (PM), small white (SW), and small yellow (SY) follicles in hens with low and high egg-laying capacities to explain regulatory pathways influencing reproductive outcomes. Specific gene expression patterns were observed that correlated with follicular growth, steroidogenesis, and granulosa cell proliferation. Heatmap clustering and principal component analysis revealed transcriptional divergence between low- and high-laying hens, suggesting that coordinated changes in signaling pathways influence egg-laying performance. High-laying hens intricated an upregulation of the PI3K-AKT-FOXO3, TGF-β, and Wnt/β-catenin pathways, which facilitate early follicular development, granulosa cell proliferation, and folliculogenesis. Higher phosphorylation of AKT and reduced nuclear FOXO3 activity were associated with enhanced primordial follicle growth. Increased TGF-β signaling, as demonstrated by higher levels of SMAD2/3/4 and cell cycle regulators, promoted granulosa cell proliferation in primary follicles (PMF). In SWF, higher levels of β-catenin and its downstream genes, such as c-Myc and cyclin D1, promoted follicle development. High-laying hens revealed increased expression of FSHR, CYP19A1, 17β-HSD, CYP1A1, and CYP1B1 in SYF, signifying enhanced FSH level and steroidogenesis. Similarly, low-laying hens exhibited downregulation of key genes, suggesting reduced follicular development and hormone signaling. These findings identify key regulatory networks and molecular markers associated with reproductive performance, providing targets for genetic selection and interventions to enhance egg production while reducing the risk of hormonal overstimulation. Full article
(This article belongs to the Special Issue Advances in Genetic Analysis of Important Traits in Poultry)
Show Figures

Figure 1

18 pages, 10717 KB  
Article
Evodiamine Inhibits Colorectal Cancer by Downregulating ASS1 via Wnt/β-Catenin/c-MYC Pathway to Block Arginine Synthesis
by Huimin Wang, Hao Deng, Jiaming He, Jing Ma, Yunying Li, Haoyue Lv, Jianhua Ran, Dilong Chen and Jing Li
Pharmaceuticals 2025, 18(11), 1736; https://doi.org/10.3390/ph18111736 - 14 Nov 2025
Abstract
Background: Argininosuccinate synthase 1 (ASS1), a key enzyme in arginine biosynthesis, is highly expressed in colorectal cancer (CRC) and promotes cancer progression, making it a potential therapeutic target. Evodiamine (EVO), a natural alkaloid from Evodia rutaecarpa acts as a novel Wnt signaling [...] Read more.
Background: Argininosuccinate synthase 1 (ASS1), a key enzyme in arginine biosynthesis, is highly expressed in colorectal cancer (CRC) and promotes cancer progression, making it a potential therapeutic target. Evodiamine (EVO), a natural alkaloid from Evodia rutaecarpa acts as a novel Wnt signaling pathway inhibitor with strong anticancer activity against various cancers. However, its exact therapeutic mechanism in CRC remains unclear. Methods: To address this gap, experiments included enzyme-linked immunosorbent assay (ELISA) to test EVO’s effect on CRC arginine production; CCK-8, EdU, colony formation, and wound-healing assays to assess CRC cell proliferation and migration; RT-qPCR, Western blot, immunofluorescence (IF), and ShASS1 for mechanism exploration and target validation; and a syngeneic tumor allograft model to study EVO’s metabolic regulation and anticancer efficacy in CRC. Results: In vitro, EVO significantly inhibited arginine synthesis metabolism and reduced CRC cell proliferation/migration. In vivo, it suppressed tumor tissue arginine metabolism, slowed allograft tumor growth, and decreased ASS1 expression. Mechanistically, EVO concentration-dependently reduced ASS1 via the Wnt/β-catenin/c-MYC pathway; ShASS1 replicated EVO’s anticancer effects, confirming ASS1’s mediating role. Conclusions: EVO downregulates ASS1 via the Wnt/β-catenin/c-MYC pathway disrupts CRC arginine synthesis metabolism and inhibits CRC cell proliferation/migration. These results support the interaction between metabolic regulation and signaling pathways, highlighting EVO as a promising CRC therapeutic candidate. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

28 pages, 2981 KB  
Article
RYR1-Related Myopathies Involve More than Calcium Dysregulation: Insights from Transcriptomic Profiling
by Daniele Sabbatini, Domenico Gorgoglione, Giovanni Minervini, Aurora Fusto, Matteo Suman, Chiara Romualdi, Sara Vianello, Giuliana Capece, Gianni Sorarù, Caterina Marchioretti, Maria Pennuto, Luca Vedovelli, Gyorgy Szabadkai, Luca Bello and Elena Pegoraro
Biomolecules 2025, 15(11), 1599; https://doi.org/10.3390/biom15111599 - 14 Nov 2025
Abstract
Ryanodine receptor 1-related myopathies (RYR1-RM) are caused by RYR1 gene variants and comprise a wide spectrum of histopathological manifestations. Here, we focus on patients carrying RYR1 variants and muscle histopathology consistent with central core disease (CCD) or multi-minicore disease (MmD). RNA-sequencing analyses of [...] Read more.
Ryanodine receptor 1-related myopathies (RYR1-RM) are caused by RYR1 gene variants and comprise a wide spectrum of histopathological manifestations. Here, we focus on patients carrying RYR1 variants and muscle histopathology consistent with central core disease (CCD) or multi-minicore disease (MmD). RNA-sequencing analyses of skeletal muscle biopsies obtained from both CCD and MmD patients and from healthy controls were performed to better understand the molecular pathways activated by RYR1 variants. Our analyses revealed that, beyond the well-established role of RYR1 in calcium homeostasis, broader cellular pathways are implicated. In CCD, differentially expressed genes were enriched for pathways related to oxidative stress response, SMAD signalling, and apoptosis, consistent with the role of intracellular calcium dysregulation in promoting mitochondrial dysfunction and cell death. In contrast, MmD patients exhibited enrichment of pathways related to immune activation. This was corroborated by the upregulation of GTPase-regulating genes and the down-regulation of transcriptional repressors such as ZFP36 and ATN1. When considering all RYR1-RM patients collectively, Wnt signalling, immune-related pathways, and oxidative phosphorylation emerged as shared enriched pathways, indicating possible convergent mechanisms across histopathological phenotypes. Our study suggests that complex gene regulation driven by RYR1 variants may be a unifying feature in CCD and MmD, offering new insight into potential therapeutic targets. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

18 pages, 641 KB  
Review
Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance
by David Mantle
Int. J. Mol. Sci. 2025, 26(22), 11024; https://doi.org/10.3390/ijms262211024 - 14 Nov 2025
Abstract
Intracellular signalling pathways provide a mechanism to connect events at a cell surface to the nucleus and are of fundamental importance to normal cell functioning. Intracellular signalling pathways control many aspects of cell metabolism, including mitochondrial function, oxidative stress, inflammation, and apoptosis/ferroptosis. Randomised [...] Read more.
Intracellular signalling pathways provide a mechanism to connect events at a cell surface to the nucleus and are of fundamental importance to normal cell functioning. Intracellular signalling pathways control many aspects of cell metabolism, including mitochondrial function, oxidative stress, inflammation, and apoptosis/ferroptosis. Randomised controlled clinical trials supplementing coenzyme Q10 (CoQ10) have reported significant clinical improvements in a number of disorders, in turn associated with the action of CoQ10 to promote normal mitochondrial function, reduce oxidative stress and inflammation, and mediate apoptosis and ferroptosis. However, the precise mechanisms by which CoQ10 facilitates beneficial changes in the above factors is not completely understood. In the present article, the evidence we have reviewed provides a supporting rationale that the beneficial role of CoQ10 in the above disorders occurs via mediation of major intracellular signalling pathways, including the Nrf2/NQO1, NF-κB, P13/AKT/mTOR, MAPK, JAK/STAT, WNT/B-catenin, AMPK-YAP-OPA1, and hedgehog (Hh) pathways; the clinical consequences of such mediation are also reviewed. Full article
(This article belongs to the Special Issue Signalling Pathways in Metabolic Diseases and Cancers)
Show Figures

Figure 1

22 pages, 9779 KB  
Article
Dietary Polyphenol Combinations Have a Multifaceted Inhibitory Effect on Metabolic Rewiring and Signaling Pathways in Neuroblastoma
by Natalia Karpova, Elizaveta Fefilova, Alexandra Daks, Sergey Parfenyev, Alexander Nazarov, Nick A. Barlev and Oleg Shuvalov
Pharmaceuticals 2025, 18(11), 1717; https://doi.org/10.3390/ph18111717 - 12 Nov 2025
Viewed by 98
Abstract
Background/Objectives: Numerous studies have demonstrated that dietary plant-derived polyphenols suppress signaling and metabolic pathways in various malignancies, including neuroblastoma. In the present study, we compared the inhibitory activities of selected polyphenols and their combinations on key metabolic and signaling pathways in two [...] Read more.
Background/Objectives: Numerous studies have demonstrated that dietary plant-derived polyphenols suppress signaling and metabolic pathways in various malignancies, including neuroblastoma. In the present study, we compared the inhibitory activities of selected polyphenols and their combinations on key metabolic and signaling pathways in two human neuroblastoma cell lines and two noncancerous cell lines—mesenchymal stem cells (MSCs). Methods: The influence of polyphenols on neuroblastoma cells and MSCs were studied via an MTT-assay, cell cycle analysis, and an apoptosis assay (flow cytometry). Chou-Talalay algorithms were used to quantify drug interactions. SeaHorse energy profiling was applied to study energy metabolism. The influence of the compounds on metabolic enzymes and signaling pathways was examined using immunoblotting. Total protein biosynthesis was assessed using o-propargyl-puromycin labeling (flow cytometry). Results: While most of the studied polyphenols displayed a more significant inhibitory effect on neuroblastoma cells than on mesenchymal stem cells (MSCs), we found that the combinations of curcumin and quercetin (CQ) and curcumin, quercetin, and resveratrol (CQR) were significantly superior to the individual compounds. These combinations displayed synergistic effects and inhibited the cell cycle while inducing apoptosis. The CQ and CQR combinations effectively suppressed metabolic reprogramming by downregulating key enzymes of glycolysis, respiration, one-carbon metabolism, glutaminolysis, and fatty acid biosynthesis, as well as N-Myc and c-Myc, which are master regulators of metabolic processes. Furthermore, CQ and CQR inhibited AKT/mTOR, MAPK/ERK, and WNT/β-catenin signaling pathways and total protein biosynthesis and sensitized malignant cells to doxorubicin. Conclusions: Polyphenol combinations exert multifaceted inhibitory effects on metabolic rewiring and signaling networks in neuroblastoma cells. Full article
Show Figures

Graphical abstract

16 pages, 1514 KB  
Article
Periostin-Induced Wnt10a Activation Promotes Dental Pulp Stem Cell Migration During Pulp Regeneration
by Keisuke Nakamura, Natsuki Iida, Yuki Hayashi, Taku Futenma, Shintaro Sakatoku, Yoshihiko Sugita and Hiroyuki Nawa
Life 2025, 15(11), 1732; https://doi.org/10.3390/life15111732 - 12 Nov 2025
Viewed by 175
Abstract
Background: Periostin can be considered a stimulator of Wnt. Elucidating the relationship between Wnt10a and Periostin in dental pulp stem cells is considered necessary for a deeper understanding of the mechanisms of dental pulp regeneration. Methods: Regenerated dental pulp from ectopic root grafts [...] Read more.
Background: Periostin can be considered a stimulator of Wnt. Elucidating the relationship between Wnt10a and Periostin in dental pulp stem cells is considered necessary for a deeper understanding of the mechanisms of dental pulp regeneration. Methods: Regenerated dental pulp from ectopic root grafts was double-stained with BrdU and Wnt10a, and the positivity rates were analyzed. Furthermore, the expression levels of Wnt10a, LRP5/6, DKK1, and Periostin within the regenerated tissue were analyzed by PCR. The expression levels of Wnt10a, LRP5/6, DKK1, and Periostin in cells stimulated with Periostin were analyzed by PCR. Wnt10a protein expression was analyzed by Western blotting and ELISA. Similar evaluations were performed with co-stimulation by Periostin and DKK1(Sample size:4). In each experiment, cells not stimulated with periostin served as the control group. Statistical analysis involved confirming the normal distribution of data using QQ plots, followed by one-way analysis of variance and post hoc Turkey’s test. Results: Migrating dental pulp stem cells expressed Wnt10a, and migration was additionally inhibited by its antagonist DKK1. Furthermore, Periostin stimulation increased Wnt10a secretion and suppressed DKK1. Conclusions: Periostin significantly increased Wnt10a expression and DPSC migration, while DKK1 inhibited these effects. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

20 pages, 2851 KB  
Article
Retinal Ischemia: Therapeutic Effects and Mechanisms of Paeoniflorin
by Windsor Wen-Jin Chao, Howard Wen-Haur Chao, Pai-Huei Peng, Yi-Tzu Lee and Hsiao-Ming Chao
Int. J. Mol. Sci. 2025, 26(22), 10924; https://doi.org/10.3390/ijms262210924 - 11 Nov 2025
Viewed by 134
Abstract
Retinal ischemia is a key factor in the progression of vision-threatening ocular diseases, including central retinal artery/vein occlusion, exudative age-related macular degeneration (eAMD), and proliferative diabetic retinopathy. This study investigates the effects of paeoniflorin along with its related neuroprotective molecular pathways in the [...] Read more.
Retinal ischemia is a key factor in the progression of vision-threatening ocular diseases, including central retinal artery/vein occlusion, exudative age-related macular degeneration (eAMD), and proliferative diabetic retinopathy. This study investigates the effects of paeoniflorin along with its related neuroprotective molecular pathways in the treatment of retinal ischemia. Free radical or ischemic-like damage was induced by incubating retinal pigment epithelium (RPE) cells for 24 h with 1 mM hydrogen peroxide (H2O2) or by subjecting retinal neuronal cells to 8 h of oxygen–glucose deprivation (OGD). Both treatments caused significant cell loss. Treatment with paeoniflorin significantly increased cell viability at 0.5 mM in both cell types. In a Wistar rat model of retinal ischemia and reperfusion (I/R) elicited by sustained high intraocular pressure (HIOP), pre-treatment with 0.5 mM paeoniflorin mitigated the ischemia-induced decline in ERG b-wave amplitude, reduction in whole and inner retinal thickness, loss of fluorogold-labeled retinal ganglion cells, and formation of apoptotic cells. Meanwhile, paeoniflorin effectively downregulated pro-neovascular mediators β-catenin, hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF), and the pro-inflammatory/angiogenic biomarker angiopoietin-2 (Ang-2), producing effects similar to the Wnt/β-catenin inhibitor (dickkopf-related protein 1), anti-angiogenic pigment epithelium-derived factor (PEDF), and anti-VEGF Avastin (bevacizumab). These findings suggest that paeoniflorin may protect against retinal ischemia through its anti-inflammatory, anti-neovascular/angiogenic, antioxidative, and neuroprotective properties. Full article
(This article belongs to the Special Issue Molecular Insight into Retinal Diseases)
Show Figures

Figure 1

15 pages, 40390 KB  
Article
Fisetin Inhibits Periodontal Pathogen-Induced EMT in Oral Squamous Cell Carcinoma via the Wnt/β-Catenin Pathway
by Ruoyao Zhang, Hiroki Takigawa, Hugo Maruyama, Takayuki Nambu, Chiho Mashimo and Toshinori Okinaga
Nutrients 2025, 17(22), 3522; https://doi.org/10.3390/nu17223522 - 11 Nov 2025
Viewed by 189
Abstract
Objective: Previous reports showed that periodontopathic bacteria induce epithelial–mesenchymal transition (EMT) in oral squamous cell carcinoma (OSCC). Fisetin, a foodborne flavonoid, is reportedly associated with anticancer potential in various carcinogenic processes. This study aimed to elucidate the effects of fisetin on Fusobacterium [...] Read more.
Objective: Previous reports showed that periodontopathic bacteria induce epithelial–mesenchymal transition (EMT) in oral squamous cell carcinoma (OSCC). Fisetin, a foodborne flavonoid, is reportedly associated with anticancer potential in various carcinogenic processes. This study aimed to elucidate the effects of fisetin on Fusobacterium nucleatum- and Porphyromonas gingivalis-induced EMT in OSCC cells. Methods: OSCC cells were co-cultured with live and heat-killed forms of F. nucleatum and P. gingivalis. The concentration of fisetin was set at 10 μM. Morphological changes in the OSCC cells were observed under a light microscope. Cell viability was measured using the Cell Counting Kit-8 assay, whereas migration was examined via wound healing. The mRNA expression of EMT-related markers was quantified using quantitative real-time polymerase chain reaction (PCR), and the expression of EMT-related markers and Wnt pathway-associated proteins was examined via Western blotting. Results: At a multiplicity of infection (MOI) of 300:1 for F. nucleatum and 100:1 for P. gingivalis, OSCC cell viability remained unchanged; however, wound closure rates increased significantly relative to the control. Likewise, treatment with fisetin (10 µM) did not materially alter viability; nevertheless, it attenuated promigratory effects induced by heat-killed periodontal pathogens at 3 h and 6 h. The OSCC cells exhibited EMT-like morphological changes after 6 h of co-culture with heat-killed pathogens. Consistently, reverse-transcriptase quantitative PCR and Western blot analyses showed increased expression of TWIST, ZEB1, and N-cadherin, accompanied by decreased E-cadherin expression, which was more pronounced in F. nucleatum than in P. gingivalis. However, fisetin reversed these trends. Moreover, co-culture with heat-killed pathogens markedly elevated β-catenin protein levels. In line with modulation of canonical Wnt/β-catenin signaling, fisetin and a Wnt inhibitor reduced β-catenin expression, whereas co-treatment with a Wnt agonist restored β-catenin levels in the presence of fisetin. Conclusions: Heat-killed F. nucleatum and P. gingivalis induced EMT in OSCC cells, with F. nucleatum exerting the strongest effect. Fisetin suppressed pathogen-driven EMT, at least partly via canonical Wnt/β-catenin signaling, highlighting its potential therapeutic value and warranting further investigation. Full article
Show Figures

Graphical abstract

13 pages, 2829 KB  
Article
Gene Expression Profiling Provides an Improved Characterization of CD79B-Mutated Diffuse Large B-Cell Lymphomas
by Luis Grossmann, Wolfgang Jagla, Marcus Bettstetter, Simone Bertz, Stephan Schwarz-Furlan, Thomas Richter, Tobias Dechow, Thomas Decker, Martin Dreyling, Karl Sotlar, Harald Bartsch, Arndt Hartmann, Julius Honecker and Andreas Gaumann
J. Pers. Med. 2025, 15(11), 548; https://doi.org/10.3390/jpm15110548 - 10 Nov 2025
Viewed by 136
Abstract
Background and Objectives: Diffuse large B-cell lymphomas (DLBCLs) are heterogeneous neoplasms. CD79B and MYD88 mutations are associated with the activated B-cell-like (ABC) subtype of DLBCL and often co-occur and lead to constitutive activation of the NF-κB pathway. Several different genetic classifications to [...] Read more.
Background and Objectives: Diffuse large B-cell lymphomas (DLBCLs) are heterogeneous neoplasms. CD79B and MYD88 mutations are associated with the activated B-cell-like (ABC) subtype of DLBCL and often co-occur and lead to constitutive activation of the NF-κB pathway. Several different genetic classifications to date have recognized CD79B- and MYD88-mutated DLBCLs as a unique subtype with poor response to therapy and unfavorable survival. However, little is known about gene expression in DLBCLs with mutated CD79B (and MYD88) in comparison to their wild type counterparts. The objective of this study was to compare the gene expression in DLBCLs according to their CD79B mutational status. Methods: A total of 48 primary, treatment-naïve DLBCLs (CD79B-mutated: 35%/n = 17, CD79B-wild type: 65%/n = 31) were investigated using RNA expression profiling (770 genes), followed by immunohistochemical analysis of the up-regulated genes and survival analysis. Results: The gene expression analysis revealed that downstream of CD79B CARD11 and the NF-κB targets NFKBIZ, IL10, IL12A, PIM1 and BCL2A1 were up-regulated in CD79B-mutated DLBCLs. The strongest up-regulation was detected for ARNT2 and WNT11. Other up-regulated genes included the apoptosis-related BID and granzyme B, as well as genes of cell cycle regulation such as RUNX1, RUNX1T1 and RASGRF1. Up-regulation was also found for IL7, STAT3, MLLT4, CD14 and the HSP90B1 subunit. TP53 mutation showed an association with poorer overall survival in a secondary analysis, consistent with prior reports, while survival by CD79B/MYD88 mutation status and the differentially expressed genes showed no significant differences in this cohort. Conclusions: In conclusion, the current study identified novel up-regulated genes in CD79B-mutated DLBCLs beyond NF-κB pathway signaling, which may contribute to a better definition of potential therapeutic targets and further improves the characterization of this distinct and aggressive DLBCL subgroup. Full article
(This article belongs to the Special Issue Molecular Pathology in Cancer Research)
Show Figures

Figure 1

23 pages, 3176 KB  
Article
In Silico Analysis of Serum Albumin Binding by Bone-Regenerative Hyaluronan-Based Molecules
by Pauline Kramp, Aydin Özmaldar, Gloria Ruiz-Gómez and M. Teresa Pisabarro
Pharmaceutics 2025, 17(11), 1445; https://doi.org/10.3390/pharmaceutics17111445 - 8 Nov 2025
Viewed by 266
Abstract
Background: The binding of glycosaminoglycans (GAG) to Wnt signaling components plays a key regulatory role in bone formation and regeneration. We previously reported de novo designed chemically modified hyaluronan derivatives, named REGAG (Rationally Engineered GAG), which demonstrated bone-regenerative properties in a mouse [...] Read more.
Background: The binding of glycosaminoglycans (GAG) to Wnt signaling components plays a key regulatory role in bone formation and regeneration. We previously reported de novo designed chemically modified hyaluronan derivatives, named REGAG (Rationally Engineered GAG), which demonstrated bone-regenerative properties in a mouse calvaria defect model. To gain initial insights into the pharmacological profile of two REGAG currently under preclinical investigation in mice, we performed a comprehensive in silico investigation of their binding to human and murine serum albumin (HSA and MSA), as it might influence their ADME properties. Furthermore, we evaluated whether REGAG binding might impact the recognition of well-characterized HSA-binding drugs. Methods: State-of-the-art in silico ADMET tools, docking and molecular dynamics simulations were used to predict and characterize the interaction of REGAG with HSA and MSA, and to investigate the molecular mechanisms involved at the atomic level. Results: The investigated REGAG molecules show a consistent binding preference for the FA1 site in both proteins, and an additional preference for the FA7 site in HSA. Their recognition might induce protein conformational changes and alter the functional state. Furthermore, REGAG’s conformational adaptability is predicted to influence their binding to the FA5/6 and FA8/9 sites of HSA, and to the FA3/4 and FA7 sites of MSA. Conclusions: Our investigations predict the binding of two hyaluronan derivatives to HSA and MSA. The mechanistic insights gained into the molecular recognition of these two REGAG molecules offer valuable information for their potential clinical application and serve as a rational basis for future molecular design aimed at improving pharmacokinetic properties. Full article
(This article belongs to the Special Issue Hyaluronic Acid-Based Drug Delivery Systems)
Show Figures

Graphical abstract

18 pages, 2640 KB  
Article
Full-Length Transcriptome Characterization and Molecular Analysis of the Longfin Batfish (Platax teira)
by Lingeng Lv, Baosuo Liu, Huayang Guo, Kecheng Zhu, Nan Zhang, Jinhui Sun and Dianchang Zhang
Fishes 2025, 10(11), 575; https://doi.org/10.3390/fishes10110575 - 8 Nov 2025
Viewed by 145
Abstract
Platax teira is a marine fish species with both ornamental and economic value, but it faces challenges in aquaculture due to environmental stress and disease. Genetic research on P. teira has been limited due to the limitations of the partially incomplete reference genome [...] Read more.
Platax teira is a marine fish species with both ornamental and economic value, but it faces challenges in aquaculture due to environmental stress and disease. Genetic research on P. teira has been limited due to the limitations of the partially incomplete reference genome and the lack of a complete transcriptome. In this study, we utilized PacBio SMRT sequencing to generate a full-length transcriptome for P. teira, obtaining 39,770 isoforms, including 32,265 known gene-related transcripts and 4730 novel transcripts from 3455 new genes. All novel genes were annotated, and enrichment analysis revealed significant associations between immune-related pathways, such as cAMP, MAPK, PI3K-Akt, and Wnt. We also identified 14,398 alternative splicing events, 2754 alternative polyadenylation events, 42,250 SSRs, 1569 transcription factors, and 2067 long non-coding RNAs. Additionally, protein–protein interaction (PPI) analysis of immune-related pathways predicted chemokines as key immune factors among novel genes. Domain prediction analysis highlighted the diverse functional potential of immune factors such as NLRC3, tyrosine kinase 2, and A2M in different alternative splicing events. Overall, the characterization of the full-length transcriptome dataset of P. teira lays the foundation for further studies on its genetic analysis and immune regulation. Full article
(This article belongs to the Special Issue Molecular Genetics and Genomics of Marine Fishes)
Show Figures

Figure 1

12 pages, 4939 KB  
Article
Levobupivacaine Administration Suppressed Cell Metabolism in Human Adenocarcinoma A549 Cells
by Masae Iwasaki, Makiko Yamamoto, Masahiro Tomihari, Kaori Fujii and Masashi Ishikawa
Int. J. Mol. Sci. 2025, 26(22), 10833; https://doi.org/10.3390/ijms262210833 - 7 Nov 2025
Viewed by 141
Abstract
Perioperative anesthesia might directly alter cancer cell biology. We investigated the effects of levobupivacaine treatment on lung adenocarcinoma cells. A549 cells were treated with levobupivacaine at concentrations of 0.1 mM and 0.5 mM for 2 h. Transfection with angiotensin-converting enzyme 2 (ACE2) small [...] Read more.
Perioperative anesthesia might directly alter cancer cell biology. We investigated the effects of levobupivacaine treatment on lung adenocarcinoma cells. A549 cells were treated with levobupivacaine at concentrations of 0.1 mM and 0.5 mM for 2 h. Transfection with angiotensin-converting enzyme 2 (ACE2) small interfering RNA (siRNA) was performed 6 h before the levobupivacaine treatment. Cell proliferation was assessed using a cell counting kit 8 (CCK-8), and ATP synthesis was evaluated with the CellTiter-Glo® 2.0 assay at 0 and 24 h after anesthesia exposure. RT-PCR was performed to examine various biomarkers. The levobupivacaine treatment suppressed ATP synthesis without affecting cell proliferation. This was associated with the upregulation of ACE2 and the downregulation of pro-cancer biomarkers, including HIF-1α, MMP-9, and β-catenin. The anticancer effect of levobupivacaine was negated when ACE2 siRNA was introduced, and it was further suppressed when combined with levobupivacaine. The RT-PCR results indicated that the expressions of B-cell/CLL lymphoma 2 (BCL2) and wingless/integrated 1 (WNT1) were reduced after levobupivacaine treatment, but these effects were reversed with ACE2 siRNA induction. The administration of levobupivacaine suppressed A549 cell metabolism and downregulated HIF-1α, MMP-9, WNT1, EGFR, and BCL2 in an ACE2-dependent manner. Full article
Show Figures

Graphical abstract

20 pages, 5140 KB  
Review
Defective Neural Stem and Progenitor Cell Proliferation in Neurodevelopmental Disorders
by Aki Shigenaka, Eri Nitta, Tadashi Nakagawa, Makiko Nakagawa and Toru Hosoi
J. Dev. Biol. 2025, 13(4), 40; https://doi.org/10.3390/jdb13040040 - 7 Nov 2025
Viewed by 420
Abstract
Neurodevelopmental disorders (NDDs), including autism spectrum disorder, intellectual disability, and attention deficit hyperactivity disorder, are increasingly recognized as disorders of early brain construction arising from defects in neural stem and progenitor cell (NSPC) proliferation. NSPCs are responsible for generating the diverse neuronal and [...] Read more.
Neurodevelopmental disorders (NDDs), including autism spectrum disorder, intellectual disability, and attention deficit hyperactivity disorder, are increasingly recognized as disorders of early brain construction arising from defects in neural stem and progenitor cell (NSPC) proliferation. NSPCs are responsible for generating the diverse neuronal and glial lineages that establish cortical architecture and neural circuitry; thus, their expansion must be tightly coordinated by intrinsic cell cycle regulators and extrinsic niche-derived cues. Disruption of these mechanisms—through genetic mutations, epigenetic dysregulation, or environmental insults—can perturb the balance between NSPC self-renewal and differentiation, resulting in aberrant brain size and connectivity. Recent advances using animal models and human pluripotent stem cell-derived brain organoids have identified key signaling pathways, including Notch, Wnt, SHH, and PI3K–mTOR, as central hubs integrating proliferative cues, while transcriptional and chromatin regulators such as PAX6, CHD8, SETD5, and ANKRD11 govern gene expression essential for proper NSPC cycling. Furthermore, prenatal exposure to teratogens such as Zika virus infection, valproic acid, or metabolic stress in phenylketonuria can recapitulate proliferation defects and microcephaly, underscoring the vulnerability of NSPCs to environmental perturbation. This review summarizes emerging insights into the molecular and cellular mechanisms by which defective NSPC proliferation contributes to NDD pathogenesis, highlighting convergence among genetic and environmental factors on cell cycle control. A deeper understanding of these pathways may uncover shared therapeutic targets to restore neurodevelopmental trajectories and mitigate disease burden. Full article
Show Figures

Figure 1

Back to TopTop