error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
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
remove_circle_outline

Search Results (857)

Search Parameters:
Keywords = regulation of mTOR signalling pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 2583 KB  
Article
Chronic Resistance Exercise Combined with Nutrient Timing Enhances Skeletal Muscle Mass and Strength While Modulating Small Extracellular Vesicle miRNA Profiles
by Dávid Csala, Zoltán Ádám, Zoltán Horváth-Szalai, Balázs Sebesi, Kitti Garai, Krisztián Kvell and Márta Wilhelm
Biomedicines 2026, 14(1), 127; https://doi.org/10.3390/biomedicines14010127 - 8 Jan 2026
Abstract
Background: The anabolic window hypothesis suggests a limited post-exercise period for optimal nutrient uptake and utilization. Prior research indicates that miRNAs in extracellular vesicles (EVs) may regulate post-exercise adaptation by influencing protein synthesis. This study aimed to examine the effects of resistance [...] Read more.
Background: The anabolic window hypothesis suggests a limited post-exercise period for optimal nutrient uptake and utilization. Prior research indicates that miRNAs in extracellular vesicles (EVs) may regulate post-exercise adaptation by influencing protein synthesis. This study aimed to examine the effects of resistance exercise (RE) on physiological parameters and the expression and function of miRNAs transported in EVs. Methods: Twenty resistance-trained male participants (22 ± 2 years) completed a five-week RE program designed for hypertrophy. They consumed maltodextrin and whey protein based on assigned nutrient timing: immediately post-exercise (AE), three hours post-exercise (AE3), or no intake (CTRL). Body composition and knee extensor strength were assessed. Small EVs were isolated and then validated via three methods. Nanoparticle tracking analysis determined EV concentration and size, followed by pooled miRNA profiling and signaling pathway analysis. Results: Skeletal muscle mass significantly increased in AE (p = 0.001, g = 2) and AE3 (p = 0.028, g = 1), and it was higher in AE compared to CTRL (p = 0.013, η2 = 0.41), while knee extensor strength improved only in AE (p = 0.032, g = 0.9). Body fat percentage significantly decreased in all groups, AE (p = 0.005, g = 1.5), AE3 (p = 0.024, g = 1), and CTRL (p = 0.005, g = 1.7). Vesicle concentration significantly increased in the AE group (p = 0.043, r = 0.7), while it decreased in the CTRL group (p = 0.046, r = 0.8). Distinct miRNA expression profiles emerged post-intervention: 20 miRNAs were upregulated in AE, while 13 in AE3 and 15 in CTRL were downregulated. Conclusions: Nutrient timing influences training adaptation but is not more critical than total macronutrient intake. Changes in EV-transported miRNAs may regulate anabolic processes via the PI3K-AKT-mTOR and FoxO pathways through PTEN regulation. Full article
(This article belongs to the Special Issue MicroRNA and Its Role in Human Health, 2nd Edition)
Show Figures

Figure 1

13 pages, 1712 KB  
Article
Endogenous Bioelectrical Modulation by REAC Metabolic Optimization-IBZ Modulates SIRT1, PPAR-γ, and Metabolic Signaling Pathways in Human Fibroblasts
by Sara Cruciani, Vania Fontani, Arianna Rinaldi, Salvatore Rinaldi and Margherita Maioli
Cells 2026, 15(2), 106; https://doi.org/10.3390/cells15020106 - 7 Jan 2026
Abstract
Fibroblasts play a fundamental role in maintaining tissue architecture, regulating repair processes, and adapting to metabolic and inflammatory stress. Increasing evidence indicates that endogenous bioelectrical states contribute to gene expression regulation and cellular homeostasis. In this study, we investigated the effects of Radio [...] Read more.
Fibroblasts play a fundamental role in maintaining tissue architecture, regulating repair processes, and adapting to metabolic and inflammatory stress. Increasing evidence indicates that endogenous bioelectrical states contribute to gene expression regulation and cellular homeostasis. In this study, we investigated the effects of Radio Electric Asymmetric Conveyer (REAC) Metabolic Optimization–Inside Blue Zone (MO-IBZ) treatment on key regulators of stress response and metabolic control in human foreskin fibroblasts (HFF-1). Cells were exposed to nine standardized REAC MO-IBZ sessions, and changes in gene and protein expression were evaluated. Quantitative RT-PCR revealed a significant downregulation of SIRT1 and an upregulation of PPAR-γ expression in treated cells compared with untreated controls. These findings indicate molecular changes involving stress-responsive and metabolic regulatory pathways; however, they should be interpreted primarily as transcriptional signatures, as no direct functional stress-response or metabolic assays were performed. Immunofluorescence analysis showed visually increased expression of mTOR, IGF-1 receptor, and cytochrome c in REAC-treated fibroblasts, supporting a qualitative indication of activation of pathways associated with anabolic signaling, mitochondrial function, and metabolic efficiency. Taken together, these findings indicate that REAC MO-IBZ induces a coordinated molecular profile compatible with changes in cellular metabolic regulatory capacity. Within the framework of current bioelectrical literature, these changes may plausibly reflect broader regulatory adaptations; however, the present work does not provide direct measurements of bioelectrical parameters, functional metabolic activity, or epigenetic regulation, and therefore such interpretations remain speculative. These results provide descriptive mechanistic evidence supporting further investigation of REAC-based bioelectrical modulation as a potential strategy to influence cellular pathways involved in metabolic balance and tissue repair, encouraging future studies incorporating direct bioelectrical, epigenetic, and functional analyses. Full article
Show Figures

Figure 1

20 pages, 17475 KB  
Article
Betulinic Acid and Betulin Suppress Melanoma Growth by Modulating Apoptosis and Autophagy via PI3K/AKT/mTOR and MAPK Pathways
by Yingying Zhang, Meng Yuan, Quan Xu, Jun Lin and Pei Lin
Int. J. Mol. Sci. 2026, 27(2), 576; https://doi.org/10.3390/ijms27020576 - 6 Jan 2026
Abstract
Malignant melanoma (MM) is a highly invasive and metastatic form of skin cancer. Betulinic acid (BA) and betulin (BE) possess pharmacological activities such as heat-clearing, detoxification, and anti-tumor effects, with BA showing potent selective cytotoxicity against melanoma cells. However, their underlying mechanisms in [...] Read more.
Malignant melanoma (MM) is a highly invasive and metastatic form of skin cancer. Betulinic acid (BA) and betulin (BE) possess pharmacological activities such as heat-clearing, detoxification, and anti-tumor effects, with BA showing potent selective cytotoxicity against melanoma cells. However, their underlying mechanisms in MM treatment remain unclear. Herein, this study systematically evaluated the anti-melanoma effects of BA and BE via integrated network pharmacology, in vitro and in vivo assays. Network pharmacology analysis revealed that BA and BE exerted anti-MM effects mainly by regulating apoptosis, angiogenesis and autophagy through the PI3K/AKT and MAPK signaling pathways. In vitro, both BA and BE inhibited colony formation and migration of B16-F10 cells, induced apoptosis by enhancing DNA damage and upregulating apoptotic protein expression, increased autophagic activity, and reduced ATP production and mitochondrial membrane potential (ΔΨm). These effects were closely associated with the inhibition of the PI3K/AKT/mTOR and MAPK pathways. Notably, BA showed stronger inhibitory effects than BE on the migration, invasion and tube formation of HUVECs. In vivo assays further confirmed that BA significantly suppressed melanoma growth in C57BL/6J mice by blocking the PI3K/AKT/mTOR and MAPK pathways. Collectively, BA and BE inhibit B16-F10 cell proliferation through the regulation of apoptosis and autophagy, with BA showing particularly promising potential as a candidate agent for MM therapy. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

20 pages, 1443 KB  
Review
Cannabinoid Signaling and Autophagy in Oral Disease: Molecular Mechanisms and Therapeutic Implications
by Undral Munkhsaikhan, Md Ataur Rahman, Alivia Shasteen, Karima Ait-Aissa, Amal M. Sahyoun, Rajat Das Gupta, Modar Kassan, Ehsanul Hoque Apu and Ammaar H. Abidi
Int. J. Mol. Sci. 2026, 27(1), 525; https://doi.org/10.3390/ijms27010525 - 4 Jan 2026
Viewed by 158
Abstract
Autophagy is a well-preserved biological mechanism that is essential for sustaining homeostasis by degradation and recycling damaged organelles, misfolded proteins, and other cytoplasmic detritus. Cannabinoid signaling has emerged as a prospective regulator of diverse cellular functions, including immunological modulation, oxidative stress response, apoptosis, [...] Read more.
Autophagy is a well-preserved biological mechanism that is essential for sustaining homeostasis by degradation and recycling damaged organelles, misfolded proteins, and other cytoplasmic detritus. Cannabinoid signaling has emerged as a prospective regulator of diverse cellular functions, including immunological modulation, oxidative stress response, apoptosis, and autophagy. Dysregulation of autophagy contributes to pathogenesis and treatment resistance of several oral diseases, including oral squamous cell carcinoma (OSCC), periodontitis, and gingival inflammation. This review delineates the molecular crosstalk between cannabinoid receptor type I (CB1) and type II (CB2) activation and autophagic pathways across oral tissues. Cannabinoids, including cannabidiol (CBD) and tetrahydrocannabinol (THC), modulate key regulators like mTOR, AMPK, and Beclin-1, thereby influencing autophagic flux, inflammation, and apoptosis. Experimental studies indicate that cannabinoids inhibit the PI3K/AKT/mTOR pathway, promote reactive oxygen species (ROS)-induced autophagy, and modulate cytokine secretion, mechanisms that underline their dual anti-inflammatory and anti-cancer capabilities. In addition, cannabinoid-induced autophagy has been shown to enhance stem cell survival and differentiation, offering promise for dental pulp regeneration. Despite these promising prospects, several challenges remain, including receptor selectivity, dose-dependent variability, limited oral bioavailability, and ongoing regulatory constraints. A deeper understanding of the context-dependent regulation of autophagy by cannabinoid signaling could pave the way for innovative therapeutic interventions in dentistry. Tailored cannabinoid-based formulations, engineered for receptor specificity, tissue selectivity, and optimized delivery, hold significant potential to revolutionize oral healthcare by modulating autophagy-related molecular pathways involved in disease resolution and tissue regeneration. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

11 pages, 1499 KB  
Article
FMDV VP3 Induces IL-10 Expression in Porcine Macrophages via PI3K Interaction and PI3K/AKT-mTOR Pathway Activation
by Yuling Li, Zijing Guo, Yan Zhang, Li Luo, Chunsai He, Qiqi Xia, Jingyuan Zhang, Zhidong Zhang and Yanmin Li
Viruses 2026, 18(1), 66; https://doi.org/10.3390/v18010066 - 1 Jan 2026
Viewed by 227
Abstract
Foot-and-mouth disease virus (FMDV) infection elicits sustained, high-level interleukin-10 (IL-10) secretion in cattle and pigs, which correlates with lymphopenia and immunosuppression. We previously showed that macrophages are the principal source of IL-10 during FMDV infection in mice, but the viral trigger and host [...] Read more.
Foot-and-mouth disease virus (FMDV) infection elicits sustained, high-level interleukin-10 (IL-10) secretion in cattle and pigs, which correlates with lymphopenia and immunosuppression. We previously showed that macrophages are the principal source of IL-10 during FMDV infection in mice, but the viral trigger and host pathways remained unknown. In the present study, we examined whether the FMDV structural protein VP3 regulates IL-10 expression. To this end, a eukaryotic VP3 expression vector was transfected into porcine alveolar macrophages (3D4/21 cells), and IL-10 expression together with related signaling pathways was interrogated by qRT-PCR, ELISA, Western blot, co-immunoprecipitation (Co-IP), confocal microscopy, and luciferase reporter assays. The results showed that VP3 significantly increased IL-10 mRNA and protein levels (p < 0.001) in a time-dependent manner. Mechanistically, VP3 promoted phosphorylation of PI3K, AKT, and mTOR; this effect was abolished by the PI3K inhibitor LY294002, which also abrogated VP3-induced IL-10 secretion (p < 0.05). Furthermore, VP3 upregulated mRNA expression of STAT3, ATF1, and CREB (p < 0.05) and enhanced IL-10 promoter activity. The STAT3 inhibitor Stattic reduced IL-10 secretion by 22% (p < 0.05). Co-IP and confocal microscopy confirmed direct binding of VP3 to PI3K in the cytoplasm. In conclusion, FMDV VP3 induces IL-10 overexpression by directly activating the PI3K/AKT-mTOR signaling pathway, thereby elucidating a key mechanism of FMDV-induced immunosuppression. Full article
(This article belongs to the Special Issue Foot-and-Mouth Disease Virus)
Show Figures

Figure 1

21 pages, 1616 KB  
Review
The TRiC/CCT Complex at the Crossroads of Metabolism and Hypoxia in GBM: Implications for IDH-Dependent Therapeutic Targeting
by Giusi Alberti, Giuseppa D’Amico, Maria Antonella Augello, Francesco Cappello, Marta Anna Szychlinska, Celeste Caruso Bavisotto and Federica Scalia
Int. J. Mol. Sci. 2026, 27(1), 373; https://doi.org/10.3390/ijms27010373 - 29 Dec 2025
Viewed by 172
Abstract
Glioblastoma (GBM) is characterized by its unique molecular features, such as self-renewal and tumorigenicity of glioma stem cells that promote resistance, largely resulting in treatment failure. Among the molecular alterations significant to GBM biology and treatment, mutations in isocitrate dehydrogenase (IDH) have assumed [...] Read more.
Glioblastoma (GBM) is characterized by its unique molecular features, such as self-renewal and tumorigenicity of glioma stem cells that promote resistance, largely resulting in treatment failure. Among the molecular alterations significant to GBM biology and treatment, mutations in isocitrate dehydrogenase (IDH) have assumed particular relevance. IDH-mutant and IDH-wild-type tumors exhibit significantly different metabolic characteristics, clinical behavior, and therapeutic sensitivities, making IDH status a critical determinant in determining prognosis and treatment strategies for GBM. In the context of cancer, chaperones were shown to promote tumor progression by supporting malignant cells over healthy ones. While heat shock proteins (HSPs) have long been implicated in the molecular mechanisms of tumor phenotype progression, recent attention has turned to CCT (chaperonin containing TCP1), orchestrating proteostasis. The chaperonin CCT is being explored as a diagnostic and therapeutic target in many cancers, including GBM, owing to its involvement in key oncogenic signaling pathways such as Wnt, VEGF, EGFR, and PI3K/AKT/mTOR. However, its role in the GBM-tricarboxylic acid (TCA) cycle cascade is still not well understood. Therefore, the present review highlights the potential role of the CCT complex in regulating hypoxia-inducible factor (HIF) activation by modulating enzymes responsive to metabolites derived from glucose metabolism and the TCA cycle in a manner dependent on oxygen availability and IDH mutation status. Full article
(This article belongs to the Special Issue Targeting Glioblastoma Metabolism)
Show Figures

Figure 1

35 pages, 2242 KB  
Review
Systematic Exploration of Molecular Mechanisms and Natural Herbal Therapeutic Strategies for Cancer Cachexia
by Pengyu Han, Xingyu Zhou, Guomin Dong, Litian Ma, Xiao Han, Donghu Liu, Jin Zheng and Jin Zhang
Cancers 2026, 18(1), 104; https://doi.org/10.3390/cancers18010104 - 29 Dec 2025
Viewed by 544
Abstract
Cancer cachexia (CC) is a multifactorial, multi-organ syndrome characterized by systemic inflammation, metabolic dysregulation, anorexia, and progressive depletion of skeletal muscle and adipose tissue. Despite its high prevalence among patients with advanced malignancies, effective therapeutic options remain limited. Recent studies have elucidated the [...] Read more.
Cancer cachexia (CC) is a multifactorial, multi-organ syndrome characterized by systemic inflammation, metabolic dysregulation, anorexia, and progressive depletion of skeletal muscle and adipose tissue. Despite its high prevalence among patients with advanced malignancies, effective therapeutic options remain limited. Recent studies have elucidated the molecular underpinnings of CC and the therapeutic potential of natural herbs, highlighting the involvement of central nervous system regulation, adipose tissue, immune responses, gut microbiota, skeletal muscle, and disruptions in anabolic–catabolic signaling pathways such as mTOR, UPS, NF-κB, and STAT3. Persistent inflammation induces E3 ubiquitin ligases (Atrogin-1/MuRF-1) through cytokines including IL-6 and TNF-α, thereby impairing muscle homeostasis, while suppression of anabolic cascades such as IGF-1/mTOR further aggravates muscle atrophy. The limited efficacy and adverse effects of synthetic agents like megestrol acetate underscore the value of herbal therapies as safer adjunctive strategies. Botanicals such as Coicis Semen, Scutellaria baicalensis, and Astragalus demonstrate anti-inflammatory and muscle-preserving activities by modulating NF-κB, IL-6, and oxidative stress signaling. Numerous investigations indicate that these herbs downregulate MuRF-1 and Atrogin-1 expression, enhance appetite, and attenuate muscle loss, though they exhibit minimal influence on tumor suppression. While promising, current evidence remains largely preclinical and mechanistic validation is incomplete. This review consolidates contemporary insights into CC pathogenesis and the bioactivity of herbal interventions, highlighting the need for translational research to bridge preclinical findings with clinical applicability. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

20 pages, 1165 KB  
Review
Reprogramming the Apoptosis–Autophagy Axis in Glioblastoma: The Central Role of the Bcl-2:Beclin-1 Complex and Survival Signalling Networks
by Monika Christoff, Amelia Szczepańska, Joanna Jakubowicz-Gil and Adrian Zając
Cells 2026, 15(1), 53; https://doi.org/10.3390/cells15010053 - 27 Dec 2025
Viewed by 476
Abstract
Glioblastoma multiforme (GBM) exhibits remarkable resistance to therapy, mainly due to its capacity to modulate regulated cell death pathways. Among these, apoptosis and autophagy are dynamically interconnected, determining cell fate under therapeutic stress. The interaction between beclin-1 and Bcl-2 proteins may represent a [...] Read more.
Glioblastoma multiforme (GBM) exhibits remarkable resistance to therapy, mainly due to its capacity to modulate regulated cell death pathways. Among these, apoptosis and autophagy are dynamically interconnected, determining cell fate under therapeutic stress. The interaction between beclin-1 and Bcl-2 proteins may represent a key molecular switch that controls whether glioma cells undergo survival or death. This review highlights the crucial role of the Bcl-2:beclin-1 complex in controlling apoptosis–autophagy axis in GBM, emphasising how survival signalling networks, including PI3K/AKT/mTOR, Ras/Raf/MEK/ERK, and PLCγ1/PKC pathways regulated by the TrkB receptor, modulate this balance. We summarise recent insights into how these pathways coordinate the shift between apoptosis and autophagy in glioma cells, contributing to drug resistance. Furthermore, we highlight how modulating this crosstalk can sensitise GBM to conventional and emerging therapies. Integrating new concepts of cell death reprogramming and systems-level signalling analysis, we propose that targeting the Bcl-2:beclin-1 complex and its upstream regulators could overcome the adaptive plasticity of glioblastoma multiforme and open new directions for combination treatment strategies. Full article
(This article belongs to the Special Issue Cell Death Mechanisms and Therapeutic Opportunities in Glioblastoma)
Show Figures

Graphical abstract

30 pages, 2720 KB  
Review
Nutritional Regulation of Cardiac Metabolism and Function: Molecular and Epigenetic Mechanisms and Their Role in Cardiovascular Disease Prevention
by Lucia Capasso, Donato Mele, Rosaria Casalino, Gregorio Favale, Giulia Rollo, Giulia Verrilli, Mariarosaria Conte, Paola Bontempo, Vincenzo Carafa, Lucia Altucci and Angela Nebbioso
Nutrients 2026, 18(1), 93; https://doi.org/10.3390/nu18010093 - 27 Dec 2025
Viewed by 380
Abstract
Background: Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and are strongly influenced by dietary habits. Beyond caloric intake, nutrients act as molecular signals that regulate cardiac metabolism, mitochondrial function, inflammation, and epigenetic remodeling. Objectives: This review aims to synthesize [...] Read more.
Background: Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and are strongly influenced by dietary habits. Beyond caloric intake, nutrients act as molecular signals that regulate cardiac metabolism, mitochondrial function, inflammation, and epigenetic remodeling. Objectives: This review aims to synthesize current evidence on how dietary patterns and specific nutritional interventions regulate cardiac metabolism and function through interconnected molecular and epigenetic mechanisms, highlighting their relevance for cardiovascular disease prevention. Methods: A narrative review of the literature was conducted using PubMed, Scopus, and Web of Science, focusing on studies published between 2006 and 2025. Experimental, translational, and clinical studies addressing diet-induced modulation of cardiac metabolic pathways, oxidative and inflammatory signaling, epigenetic regulation, and gut microbiota-derived metabolites were included. Results: The analyzed literature consistently shows that unbalanced diets rich in saturated fats and refined carbohydrates impair cardiac metabolic flexibility by disrupting key nutrient-sensing pathways, including AMP-activated protein kinase (AMPK), proliferator-activated receptor alpha (PPARα), mammalian target of rapamycin (mTOR), and sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (SIRT1/PGC-1α), leading to mitochondrial dysfunction, oxidative stress, chronic inflammation, and maladaptive remodeling. In contrast, cardioprotective dietary patterns, such as caloric restriction (CR), intermittent fasting (IF), and Mediterranean and plant-based diets, enhance mitochondrial efficiency, redox balance, and metabolic adaptability. These effects are mediated by coordinated activation of AMPK-SIRT1 signaling, suppression of mTOR over-activation, modulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways, and favorable epigenetic remodeling involving DNA methylation, histone modifications, and non-coding RNAs. Emerging evidence also highlights the central role of gut microbiota-derived metabolites, particularly short-chain fatty acids, in linking diet to epigenetic and metabolic regulation of cardiac function. Conclusions: Diet quality emerges as a key determinant of cardiac metabolic health, acting through integrated molecular, epigenetic, and microbiota-mediated mechanisms. Targeted nutritional strategies can induce long-lasting cardioprotective metabolic and epigenetic adaptations, supporting the concept of diet as a modifiable molecular intervention. These findings provide a mechanistic rationale for integrating personalized nutrition into cardiovascular prevention and precision cardiology, complementing standard pharmacological therapies. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Diet-Associated Cardiac Metabolism)
Show Figures

Figure 1

20 pages, 5510 KB  
Article
Comparative Transcriptomics Reveals Novel and Differential Long-Noncoding RNA Responses Underlying Interferon-Mediated Antiviral Regulation in Porcine Alveolar Macrophages
by Jiuyi Li, Oluwaseun Adeyemi, Laura C. Miller and Yongming Sang
Pathogens 2026, 15(1), 35; https://doi.org/10.3390/pathogens15010035 - 26 Dec 2025
Viewed by 515
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a major threat to the global swine industry. Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of antiviral immunity, but their roles in porcine alveolar macrophages (PAMs)—the primary target of PRRSV—remain poorly characterized. This [...] Read more.
Porcine reproductive and respiratory syndrome virus (PRRSV) is a major threat to the global swine industry. Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of antiviral immunity, but their roles in porcine alveolar macrophages (PAMs)—the primary target of PRRSV—remain poorly characterized. This study presents a genome-wide analysis of lncRNA expression in PAMs stimulated with a PRRS modified live virus (MLV) vaccine and two type I interferons, IFN-α1 and IFN-ω5. Whole-transcriptome sequencing identified over 2000 differentially expressed lncRNAs, with IFN-ω5 inducing the most extensive transcriptional reprogramming. Weighted gene co-expression network analysis (WGCNA) revealed interferon-specific lncRNA-mRNA modules, and functional enrichment showed these lncRNAs are involved in key immune and metabolic pathways, including chemokine signaling, MAPK, and mTOR. Our findings establish a comprehensive landscape of lncRNA regulation in PAMs, highlighting their role in fine-tuning the antiviral responses and suggesting novel targets for interferon-based antiviral interventions against PRRSV. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

20 pages, 1731 KB  
Review
Cottonseed Protein as an Alternative Feed Ingredient for Fish: Nutritional Metabolism and Physiological Implications
by Yue Hu, Yang Xie, Youdi Tang, Jiarui Liu, Esau Mbokane, Rana Al-Sayed Dawood, Jie Luo, Debing Li and Quanquan Cao
Fishes 2026, 11(1), 10; https://doi.org/10.3390/fishes11010010 - 25 Dec 2025
Viewed by 221
Abstract
Against the backdrop of the continuous expansion of the global aquaculture industry and the growing demand for high-quality feed protein, the development of sustainable alternative protein sources to fishmeal is crucial. Cottonseed protein, particularly cottonseed protein concentrate, has emerged as a highly promising [...] Read more.
Against the backdrop of the continuous expansion of the global aquaculture industry and the growing demand for high-quality feed protein, the development of sustainable alternative protein sources to fishmeal is crucial. Cottonseed protein, particularly cottonseed protein concentrate, has emerged as a highly promising plant-based alternative raw material due to its high protein content and cost advantages. This review systematically evaluates the application effects, challenges, and mechanisms of action of cottonseed protein in fish feed. Core analysis indicates that the primary limiting factor of cottonseed protein is the antinutritional factor free gossypol. High-level replacement (typically >30%) of fishmeal can inhibit fish growth, reduce protein deposition, and impair intestinal health. These adverse effects are closely associated with the downregulation of the hepatic mTOR signaling pathway—a central regulator of protein synthesis and cell growth—shifting the organism’s energy allocation from growth to stress adaptation. Furthermore, the unique fatty acid profile of cottonseed protein may exacerbate energy metabolism imbalance. To overcome gossypol toxicity, physical, chemical, and biological detoxification technologies have been widely applied. Among these, biological methods (such as Bacillus subtilis fermentation and CotA laccase-catalyzed degradation) are particularly outstanding, not only efficiently removing gossypol (removal rate > 90%) but also degrading macromolecular proteins into more digestible and absorbable small peptides and amino acids, significantly enhancing the nutritional value of cottonseed protein. Although the application prospects for cottonseed protein are broad, gaps remain in current research, particularly concerning the deeper metabolic pathways, nutrient utilization efficiency, and long-term impacts on metabolic homeostasis of detoxified cottonseed protein in fish. Future research needs to employ molecular nutrition and multi-omics technologies to elucidate its metabolic mechanisms and optimize detoxification processes and precision feeding strategies. Glandless cottonseed varieties, which fundamentally address the gossypol issue, are considered the most transformative development direction. Through continuous technological innovation, cottonseed protein is expected to become a core feed protein ingredient promoting the sustainable development of the global aquaculture industry. Full article
(This article belongs to the Special Issue Immunology, Environment, and Nutrition of Aquatic Animals)
Show Figures

Graphical abstract

17 pages, 1451 KB  
Review
Targeting the MAPK Pathway in Cancer
by Sultan F. Kadasah
Int. J. Mol. Sci. 2026, 27(1), 214; https://doi.org/10.3390/ijms27010214 - 24 Dec 2025
Viewed by 330
Abstract
The mitogen-activated protein kinase (MAPK) signaling cascade is fundamental in regulating cellular proliferation and differentiation, cell survival and cell death via apoptosis. Disruption of the MAPK signaling cascade at any point can lead to the evasion of apoptosis and unchecked cell growth and [...] Read more.
The mitogen-activated protein kinase (MAPK) signaling cascade is fundamental in regulating cellular proliferation and differentiation, cell survival and cell death via apoptosis. Disruption of the MAPK signaling cascade at any point can lead to the evasion of apoptosis and unchecked cell growth and proliferation, leading to oncogenesis. This narrative review describes MAPK pathway dysregulation, its therapeutic targets, and resistance mechanisms. The therapeutic targeting of the MAPK pathway is complex due to the dual context-dependent roles of several kinases in the signaling cascade. Despite the therapeutic effectiveness of MAPK inhibitors, cancer cells develop chemoresistance that needs to be targeted via bypassing (c-Jun N-terminal kinases) JNK, protein kinase AKT and (mammalian target of rapamycin) mTOR signaling cascades, pairing MAPK inhibitors with multiple immune agents and targeting the MAPK pathway downstream of (extracellular signal-regulated kinase) ERK to prevent its reactivation mechanisms using combination therapies, downstream signaling regulators and (Proteolysis Targeting Chimeras) PROTACs. Additionally, MAPK-mediated regulation of ferroptosis is a novel oncological therapeutic targeting strategy for controlling tumor progression. The inhibition of the RAF/MAPK pathway results in alteration of several key regulators of ferroptosis, including SLCA11, GSH, GPX4 and NCO4A, hence affecting lipid cellular iron concentration and lipid peroxidation. Emerging therapies targeting the MAPK pathway should be designed considering crosstalk, compensatory signaling mechanism activation, the role of ferroptosis and the impact of the tumor microenvironment. Full article
Show Figures

Figure 1

20 pages, 3147 KB  
Article
Network Pharmacology and Molecular Docking Identify Medicarpin as a Potent CASP3 and ESR1 Binder Driving Apoptotic and Hormone-Dependent Anticancer Activity
by Yanisa Rattanapan, Sirinya Sitthirak, Aman Tedasen, Thitinat Duangchan, Hasaya Dokduang, Nawanwat C. Pattaranggoon, Krittamate Saisuwan and Takol Chareonsirisuthigul
Int. J. Mol. Sci. 2026, 27(1), 174; https://doi.org/10.3390/ijms27010174 - 23 Dec 2025
Viewed by 249
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecologic malignancies due to late diagnosis, rapid progression, and frequent chemoresistance. Despite advances in targeted therapy, durable responses are uncommon, underscoring the need for novel multitarget agents capable of modulating key oncogenic networks. Medicarpin, [...] Read more.
Ovarian cancer (OC) remains one of the most lethal gynecologic malignancies due to late diagnosis, rapid progression, and frequent chemoresistance. Despite advances in targeted therapy, durable responses are uncommon, underscoring the need for novel multitarget agents capable of modulating key oncogenic networks. Medicarpin, a natural pterocarpan phytoalexin, exhibits diverse pharmacological activities; however, its molecular mechanisms in OC are poorly defined. This study employed an integrative in silico framework combining network pharmacology, pathway enrichment, molecular docking, and survival analysis to elucidate medicarpin’s therapeutic landscape in OC. A total of 107 overlapping targets were identified, resulting in a dense protein–protein interaction network enriched in kinase-mediated and apoptotic signaling pathways. Ten hub genes were emphasized: CASP3, ESR1, mTOR, PIK3CA, CCND1, GSK3B, CDK4, PARP1, CHEK1, and ABL1. Gene Ontology and KEGG analyses demonstrated substantial enrichment in the PI3K–Akt/mTOR and prolactin signaling pathways. Docking revealed the stable binding of medicarpin to CASP3 (−6.13 kcal/mol) and ESR1 (−7.68 kcal/mol), supporting its dual regulation of hormonal and apoptotic processes. Although CASP3 and ESR1 expression alone lacked prognostic significance, their network interplay suggests synergistic relevance. Medicarpin exhibits multitarget anticancer potential in OC by modulating kinase-driven and hormone-dependent pathways, warranting further experimental validation. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

18 pages, 4707 KB  
Article
Aging Rewires Neuronal Metabolism, Exacerbating Cell Death After Ischemic Stroke: A Hidden Reason for the Failure of Neuroprotection
by Matvey Vadyukhin, Vladimir Shchekin, Petr Shegai, Andrey Kaprin and Grigory Demyashkin
Int. J. Mol. Sci. 2026, 27(1), 81; https://doi.org/10.3390/ijms27010081 - 21 Dec 2025
Viewed by 271
Abstract
Aging profoundly modifies neuronal responses to ischemia. We aimed to define age-dependent features of neuronal metabolism and cell death after ischemic stroke by assessing NeuN, NSE, and Caspase-3 in human cortical neurons and by comparing transcriptional activity within PI3K/Akt/mTOR and PI3K/Akt/FOXO3a pathways across [...] Read more.
Aging profoundly modifies neuronal responses to ischemia. We aimed to define age-dependent features of neuronal metabolism and cell death after ischemic stroke by assessing NeuN, NSE, and Caspase-3 in human cortical neurons and by comparing transcriptional activity within PI3K/Akt/mTOR and PI3K/Akt/FOXO3a pathways across age groups. The aim of this study was to determine age-dependent features of neuronal metabolism and cellular degradation in ischemic stroke based on immunohistochemical assessment of NeuN, NSE, and Caspase-3 markers in human cerebral cortex neurons, as well as to conduct a comparative analysis of gene expression in the PI3K/Akt/mTOR and PI3K/Akt/FOXO3a signaling pathways involved in the regulation of neuronal survival and apoptosis. For the investigation, frontal cortex autopsies from patients with ischemic stroke (n = 154; “young”, “middle” and “elderly”; death ≤7 days post-onset) were examined. Histology (hematoxylin–eosin) and Nissl staining were used for morphology and neuron counts. Multiplex immunofluorescence (NeuN, NSE, Caspase-3) quantified metabolically active and apoptotic neurons, and the percentage of Caspase-3+ among NeuN+ cells was calculated. qRT-PCR measured PIK3CA, AKT2, MTOR, and FOXO3A expression in the infarct border zone. Based on our results, neuronal density and NeuN/NSE expression declined with aging, and the fraction of Caspase-3+ among NeuN+ neurons in the penumbra rose (young 42%, middle 82%, elderly 89%). Morphologically “intact” penumbral neurons frequently lacked NeuN/NSE, revealing covert dysfunction. Young brains showed balanced activation of PI3K/Akt/mTOR and PI3K/Akt/FOXO3a, whereas elderly brains exhibited reduced Akt/mTOR activity with FOXO3A predominance, consistent with pro-apoptotic, inflammatory, and dysregulated autophagic signaling. Thus, aging markedly reduces neuronal metabolic activity and increases apoptotic death in the infarct border zone after ischemic stroke. In older patients, there is an almost complete loss of NeuN and NSE expression in penumbral neurons with robust activation of the caspase cascade, whereas younger patients retain a pool of metabolically active neurons. Age-dependent dysregulation of PI3K/Akt signaling—characterized by FOXO3a hyperactivation and mTOR suppression—further promotes apoptosis and dysregulated autophagy. These changes likely underlie the limited efficacy of standard neuroprotection in ischemic stroke and support the need for age-tailored neurotropic therapy aimed at enhancing pro-survival pathways within the infarct border zone. Full article
Show Figures

Figure 1

24 pages, 5356 KB  
Article
Effects of Dietary Supplementation with Dihydromyricetin on Hindgut Microbiota and Metabolite Profiles in Dairy Cows
by Jie Yu, Yingnan Ao, Hongbo Chen, Chenhui Liu, Tinxian Deng, Dingfa Wang, Min Xiang, Pingmin Wan and Lei Cheng
Microorganisms 2026, 14(1), 20; https://doi.org/10.3390/microorganisms14010020 - 21 Dec 2025
Viewed by 208
Abstract
High-yielding dairy cows suffer from a high metabolic load and oxidative stress, which lead to systemic inflammation and metabolic disorders, increasing the susceptibility of these cows to various production diseases. Dihydromyricetin (DMY) has demonstrated potent antioxidant and anti-inflammatory physiological functions; however, research into [...] Read more.
High-yielding dairy cows suffer from a high metabolic load and oxidative stress, which lead to systemic inflammation and metabolic disorders, increasing the susceptibility of these cows to various production diseases. Dihydromyricetin (DMY) has demonstrated potent antioxidant and anti-inflammatory physiological functions; however, research into its application in ruminants remains limited. This study investigated whether DMY supplementation is associated with the maintenance of metabolic homeostasis through the regulation of gut microbiota and metabolite profiles. A total of 14 mid-lactation Holstein dairy cows were randomly divided into two groups (n = 7 per group) and supplemented with DMY at 0 or 0.05% in their basal diet for 60 consecutive days. The effects of DMY on the blood biochemical indicators and the antioxidant capacity of the dairy cows were then determined. Alterations to the gut microbiome and the fecal and plasma metabolome were analyzed through 16S rDNA sequencing and untargeted metabolomics. The results showed that DMY significantly improved the activity of serum glutathione peroxidase (GSH-Px) and presented a trend of increasing the total antioxidant capacity (T-AOC). The abundance of multiple fiber-degrading and beneficial commensal bacteria in the gut, including Fibrobacter_succinogenes, Ruminococcus_albus, and Turicibacter, was significantly elevated by the DMY intervention, accompanied by the upregulation of 8,11,14-eicosatrienoic acid, myricetin, dihydro-3-coumaric acid, PGE1, L-leucine, nicotinuric acid, pantothenic acid, and pyruvate in the feces and plasma. Moreover, DMY supplementation notably reduced the abundance of potential pathogenic microbes, such as Chloroflexi, Deltaproteobacteria, RFP12, and Succinivibrio, and downregulated the levels of 12-hydroxydodecanoic acid, 12,13-DHOME (12,13-dihydroxy-9Z-octadecenoic acid), 16-hydroxyhexadecanoic acid, niacin, and glycerol 3-phosphate. These differential metabolites were principally enriched in the mTOR signaling pathway; pantothenate, nicotinate, and thiamine metabolism; glutathione metabolism; and glycolysis/gluconeogenesis. In summary, dietary supplementation with DMY increased the abundance of intestinal fiber-degrading bacteria and multiple metabolites with known anti-inflammatory and antioxidant properties in the feces and plasma, and was associated with alterations in metabolic pathways involving B-vitamins, amino acids, and glutathione. This suggests a potential role for DMY in supporting metabolic homeostasis in dairy cows. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

Back to TopTop