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
remove_circle_outline

Search Results (734)

Search Parameters:
Keywords = phosphoinositide

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2788 KB  
Article
Molecular Insights into the Synergistic Anticancer and Oxidative Stress–Modulating Activity of Quercetin and Gemcitabine
by Yasemin Afşin, Senem Alkan Akalın, İlhan Özdemir, Mehmet Cudi Tuncer and Şamil Öztürk
Antioxidants 2026, 15(1), 91; https://doi.org/10.3390/antiox15010091 - 10 Jan 2026
Viewed by 267
Abstract
Quercetin (Q), a bioactive flavonoid, exerts potent antioxidant and redox-modulating effects by activating the nuclear factor erythroid 2-related factor 2/antioxidant response Element (Nrf2/ARE) pathway and upregulating endogenous antioxidant defenses, including enzymatic antioxidants such as superoxide dismutase (SOD) and catalase (CAT), as well as [...] Read more.
Quercetin (Q), a bioactive flavonoid, exerts potent antioxidant and redox-modulating effects by activating the nuclear factor erythroid 2-related factor 2/antioxidant response Element (Nrf2/ARE) pathway and upregulating endogenous antioxidant defenses, including enzymatic antioxidants such as superoxide dismutase (SOD) and catalase (CAT), as well as non-enzymatic glutathione (GSH) and lipid peroxidation (MDA). Gemcitabine (Gem), a widely used antimetabolite chemotherapeutic, often shows limited efficacy under hypoxic and oxidative stress conditions driven by hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF)-mediated angiogenesis. This study investigated the redox-mediated synergistic effects of Q and Gem in MDA-MB-231 human breast cancer cells. Combination treatment significantly reduced cell viability beyond the expected Bliss value, indicating a synergistic interaction and enhanced apoptosis compared with single-agent treatments. Increased reactive oxygen species (ROS) production was accompanied by depletion of GSH and accumulation of MDA, establishing a pro-apoptotic oxidative stress environment. Q alone enhanced SOD and CAT activities, whereas the combination induced exhaustion of antioxidant defenses under oxidative load, reflecting a redox-adaptive response. Molecular analyses revealed downregulation of HIF-1α and VEGF, alongside upregulation of Bax and Caspase-3, confirming suppression of hypoxia-driven survival and activation of the intrinsic apoptotic pathway. Transcriptomic and enrichment analyses further identified modulation of oxidative stress- and apoptosis-related pathways, including phosphoinositide-3-kinase–protein kinase B/Akt (PI3K/Akt), HIF-1 and VEGF signaling. Collectively, these results indicate that Q potentiates Gem cytotoxicity via redox modulation, promoting controlled ROS elevation and apoptosis while suppressing hypoxia-induced survival mechanisms, highlighting the therapeutic potential of redox-based combination strategies against chemoresistant breast cancer. Full article
(This article belongs to the Special Issue Redox Biomarkers in Cancer)
Show Figures

Figure 1

33 pages, 1777 KB  
Review
Cancer Neuroscience: Linking Neuronal Plasticity with Brain Tumor Growth and Resistance
by Doaa S. R. Khafaga, Youssef Basem, Hager Mohamed AlAtar, Abanoub Sherif, Alamer Ata, Fayek Sabry, Manar T. El-Morsy and Shimaa S. Attia
Biology 2026, 15(2), 108; https://doi.org/10.3390/biology15020108 - 6 Jan 2026
Viewed by 542
Abstract
Brain tumors, particularly glioblastoma, remain among the most lethal cancers, with limited survival benefits from current genetic and molecular-targeted approaches. Emerging evidence reveals that beyond oncogenes and mutations, neuronal plasticity, long-term potentiation, synaptic remodeling, and neurotransmitter-driven signaling play a pivotal role in shaping [...] Read more.
Brain tumors, particularly glioblastoma, remain among the most lethal cancers, with limited survival benefits from current genetic and molecular-targeted approaches. Emerging evidence reveals that beyond oncogenes and mutations, neuronal plasticity, long-term potentiation, synaptic remodeling, and neurotransmitter-driven signaling play a pivotal role in shaping tumor progression and therapeutic response. This convergence of neuroscience and oncology has given rise to the field of cancer neuroscience, which explores the bidirectional interactions between neurons and malignant cells. In this review, we summarize fundamental principles of neuronal plasticity, contrasting physiological roles with pathological reprogramming in brain tumors. We highlight how tumor cells exploit synaptic input, particularly glutamatergic signaling, to enhance proliferation, invasion, and integration into neural circuits. We further discuss how neuronal-driven feedback loops contribute to therapy resistance, including chemoresistance, radioresistance, and immune evasion, mediated through pathways such as mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), and calcium influx. The tumor microenvironment, including astrocytes, microglia, and oligodendrocyte-lineage cells, emerges as an active participant in reinforcing this neuron-tumor ecosystem. Finally, this review explores therapeutic opportunities targeting neuronal plasticity, spanning pharmacological interventions, neuromodulation approaches (transcranial magnetic stimulation (TMS), deep brain stimulation (DBS), optogenetics), and computational/artificial intelligence frameworks that model neuron tumor networks to predict personalized therapy. Also, we propose future directions integrating connect omics, neuroinformatics, and brain organoid models to refine translational strategies. Full article
(This article belongs to the Special Issue Young Researchers in Neuroscience)
Show Figures

Graphical abstract

16 pages, 2307 KB  
Article
IL-1β Controls Proliferation, Apoptosis, and Necroptosis Through the PI3K/AKT/Src/NF-κB Pathway in Leukaemic Lymphoblasts
by Zitlal-Lin Victoria-Avila, Elba Reyes-Maldonado, María Lilia Domínguez-López, Jorge Vela-Ojeda, Aranza Lozada-Ruiz, Omar Rafael Alemán and Ruth Angélica Lezama
Biomedicines 2026, 14(1), 41; https://doi.org/10.3390/biomedicines14010041 - 24 Dec 2025
Viewed by 377
Abstract
Background: Chronic inflammation and the development of cancer are closely linked, with components that comprise the tumour microenvironment—including proinflammatory cytokines—exerting essential tumourigenic effects. These proinflammatory cytokines include IL-1β, which has been reported to be overexpressed in several cancers and shown to activate several [...] Read more.
Background: Chronic inflammation and the development of cancer are closely linked, with components that comprise the tumour microenvironment—including proinflammatory cytokines—exerting essential tumourigenic effects. These proinflammatory cytokines include IL-1β, which has been reported to be overexpressed in several cancers and shown to activate several signalling pathways. These pathways may involve kinases such as AKT (serine/threonine kinase) and Src (Proto-oncogene tyrosine-protein kinase), and have a broad capacity to activate nuclear factors, including NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells), which can regulate the transcription of genes encoding proteins such as cIAP1 (Cellular Inhibitor of Apoptosis Protein 1), Bcl-2 (B-cell lymphoma 2), and cyclin D1, thereby regulating processes like apoptosis and cell cycle inhibition. Objectives: The aim of this study was to investigate the role of IL-1β (Interleukin-1 beta) in regulating cell death and proliferation in RS4:11 leukaemic lymphoblasts via the PI3K (Phosphoinositide 3-kinase)/AKT/Src/NF-κB pathway using an in vitro experimental approach. Methods: We employed flow cytometry to determine the expression levels and phosphorylation status of various proteins; proliferation was assessed using the CCK-8 kit, and apoptosis was evaluated with the Annexin V kit. Results: Our findings indicate that the IL-1β-activated signalling pathway modulates these cellular processes in leukaemic lymphoblasts. Conclusions: We therefore conclude that IL-1β exerts significant effects on cell death and proliferation in leukaemic lymphoblasts through the PI3K/AKT/NF-κB pathway, with the study’s findings indicating that an inflammatory environment may promote such lymphoblasts to acquire neoplastic characteristics. As such, the proteins involved in the effects evaluated in this work could be considered as potential therapeutic targets for the treatment of Acute Lymphoblastic Leukaemia (ALL). Full article
Show Figures

Graphical abstract

50 pages, 1412 KB  
Review
Curcumin Rewires the Tumor Metabolic Landscape: Mechanisms and Clinical Prospects
by Dingya Sun, Dun Hu, Jialu Wang, Xin Li, Jun Peng and Shan Wang
Nutrients 2026, 18(1), 53; https://doi.org/10.3390/nu18010053 - 23 Dec 2025
Viewed by 692
Abstract
Metabolic reprogramming is a fundamental hallmark and a key driver of malignant tumors. By reshaping glucose, lipid, and amino acid metabolism, as well as mitochondrial function, it sustains the abnormal proliferation and survival of tumor cells, making it a crucial target for anti-tumor [...] Read more.
Metabolic reprogramming is a fundamental hallmark and a key driver of malignant tumors. By reshaping glucose, lipid, and amino acid metabolism, as well as mitochondrial function, it sustains the abnormal proliferation and survival of tumor cells, making it a crucial target for anti-tumor therapy. Curcumin, a natural multi-target compound, exhibits unique advantages in intervening in tumor metabolic reprogramming due to its low toxicity and broad-spectrum regulatory properties. In various tumor models, it can directly modulate the activity of key glycolytic enzymes, such as hexokinase 2, lactate dehydrogenase A, and pyruvate kinase M2, as well as transporters like glucose transporter 1. Furthermore, it inhibits the expression of proteins related to lipid metabolism, including fatty acid synthase and stearoyl-CoA desaturase 1, while also intervening in amino acid metabolic networks, such as glutaminase and branched-chain amino acid transaminase. Additionally, curcumin targets mitochondrial function and reactive oxygen species balance, creating multi-dimensional intervention effects through various pathways, including the induction of ferroptosis by regulating the SLC7A11/GPX4 axis and modulating gut microbiota metabolism. Its mechanism of action involves the synergistic regulation of key signaling pathways, including phosphoinositide 3-kinase/Akt, NF-κB, AMP-activated protein kinase, and hypoxia-inducible factor-1alpha. Furthermore, its specific effect profile demonstrates significant dependency on cell type and tumor model. This article systematically reviews the regulatory effects of curcumin on these critical metabolic processes and pathways in tumor metabolic reprogramming, revealing its molecular mechanisms in disrupting tumor growth and progression by targeting energy and biosynthetic metabolism. These findings provide a significant theoretical foundation and a preclinical research perspective for the development of natural antitumor drugs based on metabolic regulation, as well as for optimizing combination therapy strategies. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

16 pages, 1083 KB  
Article
The Role of Angiopoietin-2 in Post-Burn Pneumonia
by Mary Grace Murray, Ryan M. Johnson, Abigail B. Plum, Natalia Carbajal Garcia, Kevin E. Galicia, Alexandra Brady, Madison Kipp, Irene B. Helenowski, Madison M. Tschann, Connor Guzior, Richard P. Gonzalez, Mashkoor A. Choudhry and John C. Kubasiak
Eur. Burn J. 2026, 7(1), 1; https://doi.org/10.3390/ebj7010001 - 19 Dec 2025
Viewed by 192
Abstract
Background: Pneumonia contributes to post-burn morbidity and mortality. Understanding the mechanisms that predispose burn patients to pneumonia is crucial to both stratifying patients at increased risk and developing targeted interventions. Methods: A prospective observational study was conducted with 47 human patients who sustained [...] Read more.
Background: Pneumonia contributes to post-burn morbidity and mortality. Understanding the mechanisms that predispose burn patients to pneumonia is crucial to both stratifying patients at increased risk and developing targeted interventions. Methods: A prospective observational study was conducted with 47 human patients who sustained large burn injuries with serum collected on days 2 and 3 post-burn and assessed for Angiopoietin-1 (Ang-1) and -2 (Ang-2). C57BL/6 mice were subjected to either sham injury or a 12.5% total body surface area (TBSA) scald burn injury, and plasma and lungs were assessed. Results: Patients who developed pneumonia within 30 days of injury had higher serum Ang-2 and Ang-2/1 ratio on post-injury days 2 and 3. Similar to patient findings, we observed an increase in Ang-2 in burn mice compared to sham. Within the lungs of burn mice, we found significant increases in Tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 2 (TIE2) receptor transcript Tek, downstream mediators TNFAIP3 Interacting Protein 2 (Tnip2) and phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1), in addition to endothelial adhesion molecules intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), along with neutrophil infiltration and markers compared to sham. Conclusions: These findings suggest that burn injury increases Angiopoetin-2 and downstream signaling in the lungs, which may contribute to post-burn pulmonary dysfunction. Further studies are necessary to understand if modulating the Ang–TIE2 axis can protect against pneumonia post-burn. Full article
Show Figures

Figure 1

16 pages, 1820 KB  
Article
Protective Effects of Arecoline on LPS-Induced Neuroinflammation in BV2 Microglial Cells
by Xiangfei Zhang, Jingwen Cui, Jing Sun, Bei Fan, Fengzhong Wang and Cong Lu
Int. J. Mol. Sci. 2025, 26(24), 12097; https://doi.org/10.3390/ijms262412097 - 16 Dec 2025
Cited by 1 | Viewed by 439
Abstract
Natural alkaloids derived from edible and medicinal plants have recently gained attention as bioactive molecules capable of modulating neuroinflammatory processes. Arecoline, the major alkaloid constituent of Areca catechu L. (betel nut), is well known for its cholinergic actions, yet its direct regulatory influence [...] Read more.
Natural alkaloids derived from edible and medicinal plants have recently gained attention as bioactive molecules capable of modulating neuroinflammatory processes. Arecoline, the major alkaloid constituent of Areca catechu L. (betel nut), is well known for its cholinergic actions, yet its direct regulatory influence on microglial immune signaling has remained uncertain. In this study, murine BV2 microglial cells were employed to investigate whether arecoline could counteract lipopolysaccharide (LPS)-induced neuroinflammatory responses. Parameters including cell viability, nitric oxide (NO) production, cytokine secretion, and gene expression were assessed, and mechanistic analyses were focused on the Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathways. Non-toxic doses of arecoline (10–40 μmol/L) markedly decreased NO accumulation and reduced the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β). Western blot analysis further showed that arecoline suppressed LPS-activated microglial signaling by down-regulating TLR4, inhibiting NF-κB p65 phosphorylation, and limiting PI3K/AKT activation. Collectively, these data reveal that arecoline exerts immunomodulatory and neuroprotective effects through dual signaling regulation in microglia and may serve as a useful pharmacological tool or structural reference for elucidating microglial inflammatory regulation and for guiding the exploration of safer bioactive compounds. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Graphical abstract

29 pages, 3722 KB  
Review
Glial Cells in the Early Stages of Neurodegeneration: Pathogenesis and Therapeutic Targets
by Eugenia Ahremenko, Alexander Andreev, Danila Apushkin and Eduard Korkotian
Int. J. Mol. Sci. 2025, 26(24), 11995; https://doi.org/10.3390/ijms262411995 - 12 Dec 2025
Viewed by 1019
Abstract
Astrocytes and microglia constitute nearly half of all central nervous system cells and are indispensable for its proper function. Both exhibit striking morphological and functional heterogeneity, adopting either neuroprotective (A2, M2) or proinflammatory (A1, M1) phenotypes in response to cytokines, pathogen-associated molecular patterns [...] Read more.
Astrocytes and microglia constitute nearly half of all central nervous system cells and are indispensable for its proper function. Both exhibit striking morphological and functional heterogeneity, adopting either neuroprotective (A2, M2) or proinflammatory (A1, M1) phenotypes in response to cytokines, pathogen-associated molecular patterns (PAMPs)/damage-associated molecular patterns (DAMPs), toll-like receptor 4 (TLR4) activation, and NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling. Crucially, many of these phenotypic transitions arise during the earliest stages of neurodegeneration, when glial dysfunction precedes overt neuronal loss and may act as a primary driver of disease onset. This review critically examines glial-centered hypotheses of neurodegeneration, with emphasis on their roles in early disease phases: (i) microglial polarization from an M2 neuroprotective state to an M1 proinflammatory state; (ii) NLRP3 inflammasome assembly via P2X purinergic receptor 7 (P2X7R)-mediated K+ efflux; (iii) a self-amplifying astrocyte–microglia–neuron inflammatory feedback loop; (iv) impaired microglial phagocytosis and extracellular-vesicle–mediated propagation of β-amyloid (Aβ) and tau; (v) astrocytic scar formation driven by aquaporin-4 (AQP4), matrix metalloproteinase-9 (MMP-9), glial fibrillary acidic protein (GFAP)/vimentin, connexins, and janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling; (vi) cellular reprogramming of astrocytes and NG2 glia into functional neurons; and (vii) mitochondrial dysfunction in glia, including Dynamin-related protein 1/Mitochondrial fission protein 1 (Drp1/Fis1) fission imbalance and dysregulation of the sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Sirt1/PGC-1α) axis. Promising therapeutic strategies target pattern-recognition receptors (TLR4, NLRP3/caspase-1), cytokine modulators (interleukin-4 (IL-4), interleukin-10 (IL-10)), signaling cascades (JAK2–STAT, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositide 3-kinase–protein kinase B (PI3K–AKT), adenosine monophosphate-activated protein kinase (AMPK)), microglial receptors (triggering receptor expressed on myeloid cells 2 (TREM2)/spleen tyrosine kinase (SYK)/ DNAX-activating protein 10 (DAP10), siglec-3 (CD33), chemokine C-X3-C motif ligand 1/ CX3C motif chemokine receptor 1 (CX3CL1/CX3CR1), Cluster of Differentiation 200/ Cluster of Differentiation 200 receptor 1 (CD200/CD200R), P2X7R), and mitochondrial biogenesis pathways, with a focus on normalizing glial phenotypes rather than simply suppressing pathology. Interventions that restore neuroglial homeostasis at the earliest stages of disease may hold the greatest potential to delay or prevent progression. Given the complexity of glial phenotypes and molecular isoform diversity, a comprehensive, multitargeted approach is essential for mitigating Alzheimer’s disease and related neurodegenerative disorders. This review not only synthesizes pathogenesis but also highlights therapeutic opportunities, offering what we believe to be the first concise overview of the principal hypotheses implicating glial cells in neurodegeneration. Rather than focusing on isolated mechanisms, our goal is a holistic perspective—integrating diverse glial processes to enable comparison across interconnected pathological conditions. Full article
(This article belongs to the Special Issue Early Molecular Markers of Neurodegeneration)
Show Figures

Graphical abstract

11 pages, 2253 KB  
Case Report
Longitudinal Multimodal Assessment of Structure and Function in INPP5E-Related Retinopathy
by Andrea Cusumano, Marco Lombardo, Benedetto Falsini, Michele D’Ambrosio, Jacopo Sebastiani, Enrica Marchionni, Maria Rosaria D’Apice, Barbara Rizzacasa, Francesco Martelli and Giuseppe Novelli
Genes 2025, 16(12), 1407; https://doi.org/10.3390/genes16121407 - 26 Nov 2025
Viewed by 388
Abstract
Background: INPP5E-related retinopathy (INPP5E-RR) is a rare genetic disorder caused by biallelic pathogenic variants in the INPP5E gene, which encodes an enzyme critical for phosphoinositide signaling. While early-onset rod–cone dystrophy is a hallmark feature, detailed longitudinal data on the [...] Read more.
Background: INPP5E-related retinopathy (INPP5E-RR) is a rare genetic disorder caused by biallelic pathogenic variants in the INPP5E gene, which encodes an enzyme critical for phosphoinositide signaling. While early-onset rod–cone dystrophy is a hallmark feature, detailed longitudinal data on the phenotype are scarce. This study aims to report a 6-year longitudinal assessment of retinal structure and function in a case of non-syndromic INPP5E-RR. Methods: A 42-year-old female proband with compound heterozygous pathogenic missense variants in INPP5E (p.Arg486Cys and p.Arg378Cys) was monitored from 2019 to 2025. She underwent serial comprehensive ophthalmologic evaluations, including optical coherence tomography (OCT), fundus autofluorescence, adaptive optics transscleral flood illumination, full-field 30Hz flicker electroretinography (ERG), and macular frequency-doubling technology perimetry. Results: Over the 6-year follow-up, OCT imaging revealed a progressive decline in the ellipsoid zone (EZ) width, from 1220 µm to 720 µm (~80 µm/year), and in the inner nuclear layer (INL) thickness. The central outer nuclear layer (ONL) thickness was preserved, but intraretinal cysts developed. Functional testing revealed a progressive decline in cone flicker ERG amplitudes, while visual acuity and macular perimetry remained stable. Conclusions: In this genotypically confirmed case, the longitudinal data identify EZ width, INL thickness, and cone flicker ERG as robust biomarkers of disease progression in INPP5E-RR. These parameters are ideal candidates for monitoring therapeutic outcomes in future clinical trials. Full article
(This article belongs to the Special Issue Current Advances in Inherited Retinal Disease)
Show Figures

Figure 1

19 pages, 880 KB  
Review
Lipid–Protein Interplay in the Regulation of Receptor Tyrosine Kinases
by Mattia Domenichini, Anna Gogna, Camilla Maggi, Elisa Moreschi, Anna Ventura, Martina Codibue, Elisabetta Grillo, Michela Corsini and Stefania Mitola
Cells 2025, 14(23), 1836; https://doi.org/10.3390/cells14231836 - 21 Nov 2025
Viewed by 1023
Abstract
Receptor tyrosine kinases (RTKs), a class of membrane proteins involved in several physiological processes such as growth, survival, angiogenesis, and differentiation, are profoundly influenced by the microenvironment, particularly by surrounding lipids. Lipids coordinate RTK life cycle at multiple steps. First, receptor lipidation is [...] Read more.
Receptor tyrosine kinases (RTKs), a class of membrane proteins involved in several physiological processes such as growth, survival, angiogenesis, and differentiation, are profoundly influenced by the microenvironment, particularly by surrounding lipids. Lipids coordinate RTK life cycle at multiple steps. First, receptor lipidation is a key post-translational modification for receptor-targeting localization. Then, RTK dimerization and activation are regulated by membrane-enriched lipids like phosphatidylserine and phosphoinositides, gangliosides, and Cholesterol, which directly engage RTK juxtamembrane domain or cytoplasmic tail. Eventually, lipids spatially organize RTK signaling within Cholesterol- and sphingolipid-enriched membrane microdomains. These membrane rafts act as dynamic “signalosomes” coordinating receptor clustering, endocytosis, and recycling. Perturbations in lipid composition remodel raft architecture and alter RTK behavior, contributing to pathological conditions such as cancer, metabolic, and neurodegenerative disorders. Emerging lipid-targeted therapies offer a promising way to enhance RTK-directed therapies. This review aims to explore how specific lipid species and membrane domains modulate RTK activation, clustering, and endocytic recycling. By bridging biochemical and pathological perspectives, we discuss how membrane lipid composition reshapes RTK signaling in physiology and pathology, pointing to emerging opportunities for lipid-focused therapeutic modulation. Full article
Show Figures

Graphical abstract

21 pages, 10848 KB  
Article
S100 Calcium-Binding Protein P and Cathepsin E as Key Mediators in Pancreatic Cancer Tumorigenesis
by Yu Meng, Qian Deng, Ye Zhang, Fang Wei, Jun Wu and Haijiao Yan
Biomedicines 2025, 13(11), 2780; https://doi.org/10.3390/biomedicines13112780 - 14 Nov 2025
Cited by 1 | Viewed by 2472
Abstract
Background/Objectives: Pancreatic cancer (PC) remains one of the deadliest malignancies, with challenges that hinder early detection and few actionable molecular targets. In this study, we aimed to identify biomarkers predictive of PC to support its diagnosis and treatment. Methods: Proteins from formalin-fixed, paraffin-embedded [...] Read more.
Background/Objectives: Pancreatic cancer (PC) remains one of the deadliest malignancies, with challenges that hinder early detection and few actionable molecular targets. In this study, we aimed to identify biomarkers predictive of PC to support its diagnosis and treatment. Methods: Proteins from formalin-fixed, paraffin-embedded pooled samples of PC (n = 15; 5 pools) and chronic pancreatitis (n = 10; 5 pools) tissues were analyzed via label-free quantitative proteomics using liquid chromatography-tandem mass spectrometry. Immunohistochemistry (IHC) was performed on PC tissue microarrays to assess S100 calcium-binding protein P (S100P) and cathepsin E (CTSE) expression (IHC evaluable pairs: n = 78 for S100P; n = 82 for CTSE). Transwell invasion assays were conducted to evaluate the effects of these proteins on PC cell invasiveness, and Western blotting was used to validate protein expression and elucidate associated molecular mechanisms. Results: Both S100P and CTSE were overexpressed in PC tissues compared with those in adjacent normal tissues. Elevated S100P expression correlated with poor prognosis, whereas higher CTSE expression predicted favorable outcomes; both served as independent prognostic factors in PC. Functionally, S100P promoted PC cell invasion, whereas CTSE suppressed it. Mechanistically, both proteins appeared to regulate epithelial–mesenchymal transition (EMT) and invasive capacity through activation or inhibition of the phosphoinositide 3-kinase (PI3K)–protein kinase B (AKT) signaling pathway. Conclusions: Elevated expression of S100P and CTSE in PC tissues serves as independent indicators in our model of patient survival. Both proteins regulate EMT and invasion, potentially via the PI3K–AKT pathway, and hold significant promise as prognostic biomarkers and therapeutic targets in PC. Full article
Show Figures

Figure 1

1967 KB  
Proceeding Paper
Discovery of a Selective PI3K Inhibitor Through Structure-Based Docking and Multilevel In Silico Validation
by Manjiri Bharasakare, Rahul D. Jawarkar, Pravin N. Khatale and Pramod V. Burakle
Chem. Proc. 2025, 18(1), 124; https://doi.org/10.3390/ecsoc-29-26881 - 12 Nov 2025
Viewed by 62
Abstract
Phosphoinositide 3-kinase (PI3K) represents a pivotal therapeutic target implicated in cellular proliferation, metabolic processes, and oncogenic mechanisms. This research delineates a comprehensive in silico methodology for identifying effective, pharmacokinetically favorable PI3K inhibitors. Structure-based molecular docking was executed targeting the ATP-binding pocket of PI3K, [...] Read more.
Phosphoinositide 3-kinase (PI3K) represents a pivotal therapeutic target implicated in cellular proliferation, metabolic processes, and oncogenic mechanisms. This research delineates a comprehensive in silico methodology for identifying effective, pharmacokinetically favorable PI3K inhibitors. Structure-based molecular docking was executed targeting the ATP-binding pocket of PI3K, revealing that the highest-ranked compound, MOL ID: 11325, demonstrated a significant binding affinity, reflected by a docking score of −8.558 kcal/mol. ADMET and SwissADME profiling confirmed that molecule 11325 is Lipinski-compliant, P-gp non-substrate, has a bioavailability score of 0.55, no PAINS or Brenk alerts, and a favorable synthetic accessibility (2.68), supporting its drug-likeness and development potential. A 100 ns molecular dynamics simulation confirmed the stability of the PI3K–ligand complex, demonstrating minimal deviations in root mean square deviation (RMSD) and root mean square fluctuation (RMSF). The binding free energy, determined through the MMGBSA method, exhibited a favorable value (ΔG_bind ≈ −58.6 kcal/mol), thereby corroborating the ligand’s affinity. The FEL analysis revealed distinct low-energy states, while the PCA indicated minimal structural fluctuations, confirming a stable and specific binding mode. Molecule 11325 was designated as a novel, drug-like, and dynamically stable PI3K inhibitor by this integrated computational approach, indicating that it requires additional preclinical validation for therapeutic development. Full article
Show Figures

Figure 1

14 pages, 2035 KB  
Article
Extracellular ATP Suppresses Perlecan Core Protein Synthesis via P2Y2 Receptor-Mediated Inhibition of Akt Signaling in Cultured Vascular Endothelial Cells
by Lihito Ikeuchi, Takato Hara, Kazuki Kitabatake, Fumiaki Uchiumi, Chika Yamamoto, Mitsutoshi Tsukimoto, Tomoya Fujie and Toshiyuki Kaji
Int. J. Mol. Sci. 2025, 26(22), 10973; https://doi.org/10.3390/ijms262210973 - 12 Nov 2025
Viewed by 499
Abstract
Perlecan, a major heparan sulfate proteoglycan in the vascular basement membrane, plays an essential role in maintaining endothelial barrier integrity, regulating fibroblast growth factor-2 signaling, and exerting anticoagulant activity. Although alterations in perlecan expression are implicated in the initiation and progression of atherosclerosis, [...] Read more.
Perlecan, a major heparan sulfate proteoglycan in the vascular basement membrane, plays an essential role in maintaining endothelial barrier integrity, regulating fibroblast growth factor-2 signaling, and exerting anticoagulant activity. Although alterations in perlecan expression are implicated in the initiation and progression of atherosclerosis, the upstream regulatory mechanisms remain unclear. In this study, we investigated the effects of extracellular ATP on perlecan expression in vascular endothelial cells. ATP, but not ADP or adenosine, suppressed perlecan expression at both mRNA and protein levels in a time- and concentration-dependent manner. This suppression was recovered by knockdown of P2Y2 receptor (P2Y2R), but not by P2X4 receptor, P2X7 receptor, or P2Y1 receptor knockdown, indicating the selective involvement of P2Y2R. Mechanistically, ATP reduced Akt phosphorylation mediated by P2Y2R, and inhibition of Akt by inhibitors decreased perlecan expression, whereas inhibitors of phosphoinositide 3-kinase, mammalian target of rapamycin complex 1, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, c-Jun N-terminal kinases did not exhibit this recovery effect. These results suggest that ATP downregulates perlecan synthesis via the P2Y2R-mediated inhibition of Akt signaling. Given that ATP is markedly elevated under pathological conditions, such as inflammation and platelet activation, suppression of perlecan synthesis is an important mechanism by which ATP promotes vascular disease progression. Full article
(This article belongs to the Collection Feature Papers Collection in Biochemistry)
Show Figures

Graphical abstract

18 pages, 4575 KB  
Article
β-Sitosterol Enhances the Anticancer Efficacy of Oxaliplatin in COLO-205 Cells via Apoptosis and Suppression of VEGF-A, NF-κB-p65, and β-Catenin
by Sahar Khateeb, Fahad M. Almutairi, Adel I. Alalawy, Amnah Obidan, Mody Albalawi, Rehab Al-Massabi, Hanan Abdulrahman Sagini, Samah S. Abuzahrah and Eman F. S. Taha
Int. J. Mol. Sci. 2025, 26(22), 10897; https://doi.org/10.3390/ijms262210897 - 10 Nov 2025
Viewed by 548
Abstract
Colon cancer (CC) is a common malignancy characterized by poor prognostic outcomes and considerable mortality. Oxaliplatin (OXP) is commonly used in the treatment of CC; however, its efficacy may be limited by side effects and the development of resistance. β-sitosterol (β-Sit), a phytosterol [...] Read more.
Colon cancer (CC) is a common malignancy characterized by poor prognostic outcomes and considerable mortality. Oxaliplatin (OXP) is commonly used in the treatment of CC; however, its efficacy may be limited by side effects and the development of resistance. β-sitosterol (β-Sit), a phytosterol derived from plants, has been documented to be effective in the treatment of tumors. This study aimed to investigate the potential of β-Sit to enhance the antitumor efficacy of OXP in COLO-205 cells, focusing on apoptosis induction and suppression of the vascular endothelial growth factor A (VEGF-A)/survival pathway. Molecular docking studies were performed to assess the binding affinity of β-Sit with the target proteins B-cell lymphoma 2 (Bcl-2), phosphoinositide 3-kinase (PI3K), and VEGF receptor-2 (VEGFR-2). COLO-205 cells were treated with OXP, β-Sit, or a combination of OXP + β-Sit for 48 h. The combination treatment substantially lowered the IC50 achieved with 3.24 µM of OXP and 36.01 µM of β-Sit, compared to 25.64 µM for OXP alone and 275.9 µM for β-Sit alone, demonstrating a pronounced synergistic impact. The combined therapy altered the cell cycle distribution by decreasing the number of cells in the G0/G, S, and G2/M phases, coupled with an increase in the Sub-G1 population. Furthermore, apoptosis was augmented by a shift in cell death from necrosis to late apoptosis, as indicated by an increased BAX/BCL2 ratio relative to each treatment alone. Moreover, the inhibitory effect on angiogenesis was enhanced via the reduction of VEGF-A, and β-catenin and nuclear factor κB (NF-κB-p65) were suppressed, thereby preventing the growth and survival of resistant cancer cells. Additionally, molecular docking supported high binding affinities of β-Sit to Bcl-2, PI3K, and VEGFR-2. This study highlights the potential of β-Sit to enhance the anti-cancer efficacy of OXP in CC. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

31 pages, 1143 KB  
Review
Current Status of Molecularly Targeted Therapeutics in Blood Cancers
by Caitlin Kumala, Lynh Vu and Tamer E. Fandy
Int. J. Mol. Sci. 2025, 26(21), 10512; https://doi.org/10.3390/ijms262110512 - 29 Oct 2025
Viewed by 1588
Abstract
Blood cancer is characterized by the uncontrolled growth of blood cells in the bone marrow or in the lymphatic system. Chemotherapy is still considered the first line of treatment in several types of blood cancer despite its adverse effects. Recent advances in understanding [...] Read more.
Blood cancer is characterized by the uncontrolled growth of blood cells in the bone marrow or in the lymphatic system. Chemotherapy is still considered the first line of treatment in several types of blood cancer despite its adverse effects. Recent advances in understanding the pathology and genomic changes in these cancers have led to the discovery of novel drug targets and the development of new therapeutic agents. In this review, we will discuss the mechanisms of action and clinical utility of several classes of targeted therapy used in blood cancers, including inhibitors of different types of tyrosine kinase enzymes (BCR-ABL, FLT3 and BTK), BCL-2 inhibitors, phosphoinositide 3-kinase inhibitors, nuclear export inhibitors, immune therapies (monoclonal antibodies, radioimmunoconjugates, chimeric antigen receptor T-cells, immune checkpoint inhibitors, and bispecific antibodies), and proteasome-dependent drugs (proteasome inhibitors and proteolysis targeting chimeras). Further advances in identifying distinct molecular subgroups in blood cancers will offer more opportunities for novel targeted therapies and more personalized medicine approaches. Full article
Show Figures

Figure 1

21 pages, 1367 KB  
Review
Glycolytic Reprogramming in Uterine Fibroids: Genetic, Transcriptomic, Proteomic, and Metabolomic Insights
by Samya El Sayed, Alvina Pan, Valentina Vanos, Rachel Michel and Mostafa Borahay
Genes 2025, 16(11), 1268; https://doi.org/10.3390/genes16111268 - 28 Oct 2025
Viewed by 908
Abstract
Uterine leiomyomas or fibroids are a common but pernicious benign tumor impacting between 70–80% of women of reproductive age. Despite their high prevalence, the etiology of uterine fibroids is not fully understood. This review aims to highlight the distinct metabolic features that uterine [...] Read more.
Uterine leiomyomas or fibroids are a common but pernicious benign tumor impacting between 70–80% of women of reproductive age. Despite their high prevalence, the etiology of uterine fibroids is not fully understood. This review aims to highlight the distinct metabolic features that uterine fibroids adopt to meet biosynthetic demands, support proliferation, extracellular matrix production, survival, and fibrosis. Specifically, we posit the role of glycolytic reprogramming—an adaptation in fibrosis across organs (lung, kidney, heart, and liver) as a major contributor to uterine fibroid development. Previous genetic, transcriptomic, proteomic, and metabolic studies have drawn strong links between metabolism and uterine fibroid biology and identified genotype-specific metabolic alterations such as fumarate hydratase (FH) deficiency and mediator of RNA polymerase II transcription (MED12) gene mutations. Studies in non-uterine models have linked glycolysis to ECM production and fibrosis through activation of transforming growth factor-beta (TGF-β) and the canonical Wnt pathway (Wnt/β-catenin) signaling, supporting them as potential key pathways in uterine fibroid pathogenesis via glycolytic reprogramming. Other metabolic regulators, such as hypoxia-inducible factor 1-alpha (HIF-1α), mammalian target of rapamycin (mTOR), and phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), may also sustain the fibrotic phenotype through coupling signaling that drives ECM production to metabolic programming. Overall, the proposed metabolic perspective of uterine fibroid pathogenesis invites further exploration of mechanistic investigation in uterine-specific models and therapeutic targeting through larger cohort studies. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Back to TopTop