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
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 (3,161)

Search Parameters:
Keywords = PI3K → Akt Pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1853 KiB  
Review
Exploring the Protective Effects of Taxifolin in Cardiovascular Health: A Comprehensive Review
by Hwan-Hee Sim, Ju-Young Ko, Dal-Seong Gong, Dong-Wook Kim, Jung Jin Kim, Han-Kyu Lim, Hyun Jung Kim and Min-Ho Oak
Int. J. Mol. Sci. 2025, 26(16), 8051; https://doi.org/10.3390/ijms26168051 (registering DOI) - 20 Aug 2025
Abstract
Taxifolin is a natural flavonoid found in a variety of plants, including Siberian larch (Larix sibirica) and milk thistle (Silybum marianum), that has attracted attention for its multifaceted pharmacological properties, including cardioprotective effects. Through its antioxidant and anti-inflammatory activities, [...] Read more.
Taxifolin is a natural flavonoid found in a variety of plants, including Siberian larch (Larix sibirica) and milk thistle (Silybum marianum), that has attracted attention for its multifaceted pharmacological properties, including cardioprotective effects. Through its antioxidant and anti-inflammatory activities, taxifolin has shown significant therapeutic potential in cardiovascular diseases such as atherosclerosis, myocardial ischemia, and diabetic cardiomyopathy. This review highlights the cardioprotective effects of taxifolin in preclinical models of atherosclerosis, ischemia/reperfusion injury, and diabetic cardiomyopathy. Taxifolin contributes to its cardioprotective effects through key mechanisms such as modulation of pathways such as PI3K/AKT and JAK2/STAT3, inhibition of NADPH oxidase, and modulation of nitric oxide production. Recent studies have shown that taxifolin can affect glucose metabolism by modulating sodium–glucose transporter (SGLT) expression, potentially enhancing the cardioprotective effects of SGLT2 inhibitors. Given the emerging role of SGLT2 inhibitors in the management of cardiovascular disease, further investigation of the interaction of this pathway with taxifolin may provide new therapeutic insights. Although taxifolin has multifaceted potential in the prevention and treatment of cardiovascular disease, further studies are needed to better understand its mechanisms and validate its efficacy in different disease stages. This review aims to provide a rationale for the clinical application of taxifolin-based cardiovascular therapies and suggest directions for future research. Full article
(This article belongs to the Special Issue Bioactive Compounds in the Prevention of Chronic Diseases)
Show Figures

Figure 1

44 pages, 1023 KiB  
Review
Systemic Neurodegeneration and Brain Aging: Multi-Omics Disintegration, Proteostatic Collapse, and Network Failure Across the CNS
by Victor Voicu, Corneliu Toader, Matei Șerban, Răzvan-Adrian Covache-Busuioc and Alexandru Vlad Ciurea
Biomedicines 2025, 13(8), 2025; https://doi.org/10.3390/biomedicines13082025 - 20 Aug 2025
Abstract
Neurodegeneration is increasingly recognized not as a linear trajectory of protein accumulation, but as a multidimensional collapse of biological organization—spanning intracellular signaling, transcriptional identity, proteostatic integrity, organelle communication, and network-level computation. This review intends to synthesize emerging frameworks that reposition neurodegenerative diseases (ND) [...] Read more.
Neurodegeneration is increasingly recognized not as a linear trajectory of protein accumulation, but as a multidimensional collapse of biological organization—spanning intracellular signaling, transcriptional identity, proteostatic integrity, organelle communication, and network-level computation. This review intends to synthesize emerging frameworks that reposition neurodegenerative diseases (ND) as progressive breakdowns of interpretive cellular logic, rather than mere terminal consequences of protein aggregation or synaptic attrition. The discussion aims to provide a detailed mapping of how critical signaling pathways—including PI3K–AKT–mTOR, MAPK, Wnt/β-catenin, and integrated stress response cascades—undergo spatial and temporal disintegration. Special attention is directed toward the roles of RNA-binding proteins (e.g., TDP-43, FUS, ELAVL2), m6A epitranscriptomic modifiers (METTL3, YTHDF1, IGF2BP1), and non-canonical post-translational modifications (SUMOylation, crotonylation) in disrupting translation fidelity, proteostasis, and subcellular targeting. At the organelle level, the review seeks to highlight how the failure of ribosome-associated quality control (RQC), autophagosome–lysosome fusion machinery (STX17, SNAP29), and mitochondrial import/export systems (TIM/TOM complexes) generates cumulative stress and impairs neuronal triage. These dysfunctions are compounded by mitochondrial protease overload (LONP1, CLPP), UPR maladaptation, and phase-transitioned stress granules that sequester nucleocytoplasmic transport proteins and ribosomal subunits, especially in ALS and FTD contexts. Synaptic disassembly is treated not only as a downstream event, but as an early tipping point, driven by impaired PSD scaffolding, aberrant endosomal recycling (Rab5, Rab11), complement-mediated pruning (C1q/C3–CR3 axis), and excitatory–inhibitory imbalance linked to parvalbumin interneuron decay. Using insights from single-cell and spatial transcriptomics, the review illustrates how regional vulnerability to proteostatic and metabolic stress converges with signaling noise to produce entropic attractor collapse within core networks such as the DMN, SN, and FPCN. By framing neurodegeneration as an active loss of cellular and network “meaning-making”—a collapse of coordinated signal interpretation, triage prioritization, and adaptive response—the review aims to support a more integrative conceptual model. In this context, therapeutic direction may shift from damage containment toward restoring high-dimensional neuronal agency, via strategies that include the following elements: reprogrammable proteome-targeting agents (e.g., PROTACs), engineered autophagy adaptors, CRISPR-based BDNF enhancers, mitochondrial gatekeeping stabilizers, and glial-exosome neuroengineering. This synthesis intends to offer a translational scaffold for viewing neurodegeneration as not only a disorder of accumulation but as a systems-level failure of cellular reasoning—a perspective that may inform future efforts in resilience-based intervention and precision neurorestoration. Full article
(This article belongs to the Special Issue Cell Signaling and Molecular Regulation in Neurodegenerative Disease)
Show Figures

Figure 1

16 pages, 6762 KiB  
Article
Transcriptome Sequencing and Differential Analysis of Ovaries Across Diverse States (Follicular and Non-Follicular Phases)
by Jiabei Sun, Tongliang Wang, Yuheng Xue, Zhehong Shen, Chen Meng, Xinkui Yao, Jun Meng, Jianwen Wang, Hongzhong Chu, Wanlu Ren, Linling Li and Yaqi Zeng
Animals 2025, 15(16), 2436; https://doi.org/10.3390/ani15162436 - 20 Aug 2025
Abstract
The Kazakh horse, one of China’s indigenous primitive breeds, is renowned for its remarkable adaptability and distinctive physiological traits. The ovary is a vital reproductive organ in female animals, responsible for oocyte production and hormone secretion. However, limited research has been conducted on [...] Read more.
The Kazakh horse, one of China’s indigenous primitive breeds, is renowned for its remarkable adaptability and distinctive physiological traits. The ovary is a vital reproductive organ in female animals, responsible for oocyte production and hormone secretion. However, limited research has been conducted on gene expression profiles in the ovarian tissue of equine species. To address this gap, the present study performed transcriptomic sequencing on ovarian tissues from 12 Kazakh horses in different physiological states. A total of 979 differentially expressed mRNAs were identified, including 619 upregulated and 360 downregulated genes. Among these, key genes such as COL1A1, LHCGR, KISS1, NTRK2, COL1A2, and THBS4 were identified as differentially expressed. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that 374 of these genes were primarily involved in ovarian steroidogenesis, the PI3K-Akt signaling pathway, and ECM-receptor interactions among 292 enriched pathways. This study provides a comprehensive transcriptomic profile of equine ovarian tissue, offering in-depth insights into differential gene expression and signal pathways associated with ovarian development in Kazakh horses, providing theoretical foundations and referential data for future research in equine ovarian development and reproductive studies. Full article
(This article belongs to the Section Equids)
Show Figures

Figure 1

28 pages, 2605 KiB  
Review
Exercise-Induced Muscle–Fat Crosstalk: Molecular Mediators and Their Pharmacological Modulation for the Maintenance of Metabolic Flexibility in Aging
by Amelia Tero-Vescan, Hans Degens, Antonios Matsakas, Ruxandra Ștefănescu, Bianca Eugenia Ősz and Mark Slevin
Pharmaceuticals 2025, 18(8), 1222; https://doi.org/10.3390/ph18081222 - 19 Aug 2025
Abstract
Regular physical activity induces a dynamic crosstalk between skeletal muscle and adipose tissue, modulating the key molecular pathways that underlie metabolic flexibility, mitochondrial function, and inflammation. This review highlights the role of myokines and adipokines—particularly IL-6, irisin, leptin, and adiponectin—in orchestrating muscle–adipose tissue [...] Read more.
Regular physical activity induces a dynamic crosstalk between skeletal muscle and adipose tissue, modulating the key molecular pathways that underlie metabolic flexibility, mitochondrial function, and inflammation. This review highlights the role of myokines and adipokines—particularly IL-6, irisin, leptin, and adiponectin—in orchestrating muscle–adipose tissue communication during exercise. Exercise stimulates AMPK, PGC-1α, and SIRT1 signaling, promoting mitochondrial biogenesis, fatty acid oxidation, and autophagy, while also regulating muscle hypertrophy through the PI3K/Akt/mTOR and Wnt/β-catenin pathways. Simultaneously, adipose-derived factors like leptin and adiponectin modulate skeletal muscle metabolism via JAK/STAT3 and AdipoR1-mediated AMPK activation. Additionally, emerging exercise mimetics such as the mitochondrial-derived peptide MOTS-c and myostatin inhibitors are highlighted for their roles in increasing muscle mass, the browning of white adipose tissue, and improving systemic metabolic function. The review also addresses the role of anti-inflammatory compounds, including omega-3 polyunsaturated fatty acids and low-dose aspirin, in mitigating NF-κB and IL-6 signaling to protect mitochondrial health. The resulting metabolic flexibility, defined as the ability to efficiently switch between lipid and glucose oxidation, is enhanced through repeated exercise, counteracting age- and disease-related mitochondrial and functional decline. Together, these adaptations demonstrate the importance of inter-tissue signaling in maintaining energy homeostasis and preventing sarcopenia, obesity, and insulin resistance. Finally, here we propose a stratified treatment algorithm based on common age-related comorbidities, offering a framework for precision-based interventions that may offer a promising strategy to preserve metabolic plasticity and delay the age-associated decline in cardiometabolic health. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

21 pages, 967 KiB  
Review
Recent Advances in the Application of Cucurbitacin B as an Anticancer Agent
by Dongge Yin, Hongyue Chen, Shuting Lin, Yufei Sun, Xiaohong Jing, Rongrong Chang, Yang Feng, Xiaoxv Dong, Changhai Qu, Jian Ni and Xingbin Yin
Int. J. Mol. Sci. 2025, 26(16), 8003; https://doi.org/10.3390/ijms26168003 - 19 Aug 2025
Abstract
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and [...] Read more.
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and primary liver cancer. CuB exerts anticancer effects through multiple mechanisms: inducing apoptosis, cell cycle arrest (G2/M or S phase), autophagy, and cytoskeleton disruption; inhibiting migration, invasion, and angiogenesis (via VEGF/FAK/MMP-9 and Wnt/β-catenin pathways); regulating metabolic reprogramming and immune responses; inducing pyroptosis, ferroptosis, and epigenetic changes; and reversing tumor drug resistance. These effects are associated with signaling pathways like JAK/STAT, PI3K/Akt/mTOR, and FOXM1-KIF20A. To improve its application potential, strategies such as structural modification (e.g., NO donor conjugation), combination therapy (with gemcitabine or cisplatin), and nanomaterial-based delivery (e.g., liposomes and exosome-mimicking nanoparticles) have been developed to enhance efficacy, reduce toxicity, and improve bioavailability. CuB shows broad-spectrum anticancer activity, but further research is needed to clarify the mechanisms underlying its cell-specific sensitivity and interactions with the immune system. This review systematically summarizes the physicochemical properties, anticancer mechanisms, and strategies for applying CuB and suggests future research directions, providing references for scientific research and clinical translation. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Graphical abstract

16 pages, 14697 KiB  
Article
Comparative Study of Reprogramming Efficiency and Regulatory Mechanisms of Placental- and Fibroblast-Derived Induced Pluripotent Stem Cells (iPSCs) in Mules
by Fangyuan Liu, Jia Zhang, Lingyu Kong, Rihan Wu, Qiqi Jiang, Ying Lu and Xihe Li
Curr. Issues Mol. Biol. 2025, 47(8), 671; https://doi.org/10.3390/cimb47080671 - 19 Aug 2025
Abstract
As an interspecies hybrid inheriting genetic material from horse and donkey lineages, mules provide a unique model for studying allele-specific regulatory dynamics. Here, we isolated adult fibroblasts (AFs) and placental fibroblasts (PFs) from mule tissues and reprogrammed them into induced pluripotent stem cells [...] Read more.
As an interspecies hybrid inheriting genetic material from horse and donkey lineages, mules provide a unique model for studying allele-specific regulatory dynamics. Here, we isolated adult fibroblasts (AFs) and placental fibroblasts (PFs) from mule tissues and reprogrammed them into induced pluripotent stem cells (iPSCs). Intriguingly, placental fibroblast-derived iPSCs (mpiPSCs) exhibited reduced reprogramming efficiency compared to adult fibroblast-derived iPSCs (maiPSCs). Through allele-specific expression (ASE) analysis, we systematically dissected transcriptional biases in parental cell types and their reprogrammed counterparts, revealing conserved preferential expression of asinine alleles in core pluripotency regulators (e.g., POU5F1/OCT4, SOX2, NANOG) across both cell lineages. Strikingly, mpiPSCs displayed stronger asinine allele dominance than maiPSCs, suggesting tissue-specific parental genomic imprinting. Mechanistic exploration implicated PI3K-AKT signaling as a potential pathway mediating the reprogramming inefficiency in placental fibroblasts. By integrating transcriptomic profiling with ASE technology, this study uncovers allele selection hierarchies during somatic cell reprogramming in hybrids and establishes a framework for understanding how parental genomic conflicts shape pluripotency establishment. These findings advance interspecies iPSC research by delineating allele-specific regulatory networks and providing insights into the molecular constraints of hybrid cellular reprogramming. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

23 pages, 6102 KiB  
Review
Contribution and Regulation of HIF-1α in Testicular Injury Induced by Diabetes Mellitus
by Defan Wang, Zhenghong Zhang, Renfeng Xu and Zhengchao Wang
Biomolecules 2025, 15(8), 1190; https://doi.org/10.3390/biom15081190 - 19 Aug 2025
Abstract
Diabetes mellitus, as a metabolic disorder, has received growing attention for its detrimental effects on the male reproductive system (particularly the testes) manifesting as increased oxidative stress, reduced blood perfusion, heightened inflammation, and germ cell apoptosis under hyperglycemic conditions. Hypoxia-inducible factor (HIF)-1α, a [...] Read more.
Diabetes mellitus, as a metabolic disorder, has received growing attention for its detrimental effects on the male reproductive system (particularly the testes) manifesting as increased oxidative stress, reduced blood perfusion, heightened inflammation, and germ cell apoptosis under hyperglycemic conditions. Hypoxia-inducible factor (HIF)-1α, a pivotal transcription factor in cellular hypoxia responses, plays a crucial role in regulating metabolism, angiogenesis, and apoptosis. Emerging evidence underscores its significant physiological and pathological roles in diabetic testicular injury. This review outlines the structural domains, activation mechanisms, and key target genes of HIF-1α, and further examines its involvement in diabetes-induced oxidative stress, impaired perfusion, endocrine dysregulation, and the imbalance of apoptosis and autophagy in testicular tissue. Notably, HIF-1α exerts protective effects by activating canonical signaling pathways such as phosphoinositide-3 kinase (PI-3K)/protein kinase B (Akt), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), and nuclear factor (NF)-κB, thereby enhancing antioxidant gene expression, promoting angiogenesis, and upregulating anti-apoptotic proteins. Furthermore, HIF-1α may help stabilize androgen levels by preserving Leydig cell function, potentially alleviating diabetes-associated gonadal dysfunction. This review also discusses the feasibility of targeting HIF-1α as a novel therapeutic strategy. In conclusion, a comprehensive understanding of HIF-1α’s mechanistic role in diabetic testicular damage provides valuable insights into the pathogenesis of diabetes-related reproductive disorders and offers new avenues for therapeutic intervention. Full article
(This article belongs to the Special Issue Obesity, Hormones and Reproductive Health)
Show Figures

Figure 1

23 pages, 3101 KiB  
Review
Hydroxytyrosol and Brain Tumors: Mechanisms of Action and Therapeutic Potential
by Cristina Cueto-Ureña, María Jesús Ramírez-Expósito, María Pilar Carrera-González and José Manuel Martínez-Martos
Curr. Issues Mol. Biol. 2025, 47(8), 667; https://doi.org/10.3390/cimb47080667 - 18 Aug 2025
Abstract
Central nervous system (CNS) tumors, especially gliomas and IDH-wildtype glioblastoma, present high aggressiveness, low response to current treatments and limited survival. Several biological processes such as oxidative stress, inflammation, apoptosis, and autophagy are involved in their development. Hydroxytyrosol (HTX), a phenolic compound present [...] Read more.
Central nervous system (CNS) tumors, especially gliomas and IDH-wildtype glioblastoma, present high aggressiveness, low response to current treatments and limited survival. Several biological processes such as oxidative stress, inflammation, apoptosis, and autophagy are involved in their development. Hydroxytyrosol (HTX), a phenolic compound present in olives, has shown relevant effects on these processes in experimental models. This review analyzes its chemical characteristics, bioavailability, and ability to cross the blood–brain barrier, as well as its mechanisms of action. Despite its rapid metabolism, HTX can reach the brain in small but functional amounts, and various formulation methods can enhance its delivery to nervous tissue. HTX acts on cellular pathways such as Nrf2, NF-κB, JAK/STAT, PI3K/Akt and SIRT1, regulating redox balance, inflammation, programmed cell death, and autophagy. It can also influence gene expression through epigenetic mechanisms. In cell models, it has shown inhibitory effects on tumor growth and activation of apoptosis, without affecting non-tumor cells. These results support its possible usefulness as an adjunct in the treatment of brain tumors, although further studies in animal and human models are required. Full article
Show Figures

Figure 1

26 pages, 3015 KiB  
Article
Effects of Asprosin and Role of TLR4 as a Biomarker in Endometrial Cancer
by Rebecca Karkia, Cristina Sisu, Sayeh Saravi, Ioannis Kyrou, Harpal S. Randeva, Jayanta Chatterjee and Emmanouil Karteris
Molecules 2025, 30(16), 3410; https://doi.org/10.3390/molecules30163410 - 18 Aug 2025
Abstract
(1) Background: Following the discovery of the adipokine/hormone asprosin, a substantial amount of research has provided evidence for its role in the regulation of glucose homeostasis, as well as appetite, and insulin sensitivity. Its levels are dysregulated in certain disease states, including breast [...] Read more.
(1) Background: Following the discovery of the adipokine/hormone asprosin, a substantial amount of research has provided evidence for its role in the regulation of glucose homeostasis, as well as appetite, and insulin sensitivity. Its levels are dysregulated in certain disease states, including breast cancer. To date, little is known about its role in endometrial cancer (EC). The present study investigated the effects of asprosin on the transcriptome of the Ishikawa and NOU-1 EC cell lines, and assessed the expression of asprosin’s candidate receptors (TLR4, PTPRD, and OR4M1) in health and disease. (2) Methods: tissue culture, RNA extraction, RNA sequencing, reverse transcription-quantitative PCR, gene enrichment and in silico analyses were used for this study. (3) Results: TLR4 and PTPRD were significantly downregulated in EC when compared to healthy controls. TLR4 appeared to have a prognostic role in terms of overall survival (OS) in EC patients (i.e., higher expression, better OS). RNA sequencing revealed that asprosin affected 289 differentially expressed genes (DEGs) in Ishikawa cells and 307 DEGs in NOU-1 cells. Pathway enrichment included apoptosis, glycolysis, hypoxia, and PI3K/AKT/ mTOR/NOTCH signalling for Ishikawa-treated cells. In NOU-1, enriched processes included inflammatory response, epithelial-mesenchymal transition, reactive oxygen species pathways, and interferon gamma responses. Other signalling pathways included mTORC1, DNA repair, and p53, amongst others. (4) Conclusions: These findings underscore the importance of understanding receptor dynamics and signalling pathways in the context of asprosin’s role in EC, and provide evidence for a potential role of TLR4 as a diagnostic biomarker. Full article
(This article belongs to the Special Issue Novel Metabolism-Related Biomarkers in Cancer)
Show Figures

Figure 1

17 pages, 4158 KiB  
Article
Exploring the Role of Ferroptosis in the Pathophysiology and Circadian Regulation of Restless Legs Syndrome
by Maria Paola Mogavero, Giovanna Marchese, Giovanna Maria Ventola, Giuseppe Lanza, Oliviero Bruni, Luigi Ferini-Strambi and Raffaele Ferri
Biomolecules 2025, 15(8), 1184; https://doi.org/10.3390/biom15081184 - 18 Aug 2025
Abstract
The study objectives were to investigate the role of ferroptosis, the mechanism linking iron accumulation, oxidative stress, and dopaminergic dysfunction, in restless legs syndrome (RLS), and to explore its connection with circadian regulation, a key feature of RLS and a known modulator of [...] Read more.
The study objectives were to investigate the role of ferroptosis, the mechanism linking iron accumulation, oxidative stress, and dopaminergic dysfunction, in restless legs syndrome (RLS), and to explore its connection with circadian regulation, a key feature of RLS and a known modulator of ferroptosis. We conducted pathway and gene expression analyses in 17 RLS patients and 39 controls, focusing on pathways related to ferroptosis, oxidative stress, iron metabolism, dopaminergic signaling, circadian rhythms, and immune responses. Enrichment analysis, differential gene expression, and cross-pathway gene overlaps were assessed. Ferroptosis and efferocytosis pathways were significantly upregulated in RLS, while oxidative phosphorylation, phosphatidylinositol signaling, PI3K-Akt, FoxO, and adipocytokine pathways were downregulated. The circadian rhythm pathway was markedly suppressed, with 12 circadian genes downregulated, suggesting that circadian disruption may drive ferroptosis activation. Decreased expression of protective pathways, including antioxidant responses and autophagy, was associated with increased iron accumulation, oxidative stress, and inflammation. Dopaminergic synapse genes were upregulated, possibly as a compensatory response to neuronal damage. Several genes overlapped across ferroptosis, circadian, and dopaminergic pathways, indicating a shared pathogenic mechanism. Our findings support a model in which circadian disruption promotes ferroptosis in RLS, contributing to iron overload, oxidative damage, and dopaminergic dysfunction. This pathogenic cascade may also enhance immune activation and inflammation. Circadian regulation and ferroptosis emerge as promising therapeutic targets in RLS. Further studies in larger cohorts are warranted to validate these mechanistic insights. Full article
Show Figures

Graphical abstract

18 pages, 3463 KiB  
Article
Expression of miRNA in the Semitendinosus Muscle of Cattle Breeds with Varying Intramuscular Fat Deposition
by Anna Ciecierska, Abdolvahab Ebrahimpour Gorji, Alicja Majewska and Tomasz Sadkowski
Genes 2025, 16(8), 969; https://doi.org/10.3390/genes16080969 - 18 Aug 2025
Abstract
Background: This study investigates the expression of microRNAs (miRNAs) in the semitendinosus muscle of cattle breeds with varying intramuscular fat (IMF) deposition to identify key miRNA regulators of beef marbling, utilizing Hereford (HER; higher IMF) and Holstein-Friesian (HF; moderate IMF) bulls, and [...] Read more.
Background: This study investigates the expression of microRNAs (miRNAs) in the semitendinosus muscle of cattle breeds with varying intramuscular fat (IMF) deposition to identify key miRNA regulators of beef marbling, utilizing Hereford (HER; higher IMF) and Holstein-Friesian (HF; moderate IMF) bulls, and Limousin (LIM; low IMF) bulls with lower IMF in the semitendinosus muscle. Methods: MicroRNA profiling used custom bovine microarrays and the Agilent software. The selected miRNAs, miR-34a, miR-149-5p, miR-208b, miR-499, miR-660, and miR-1343-5p, were chosen for validation using real-time PCR, confirming their differential expression. Target prediction utilized miRWalk, while functional and pathway analyses were conducted using the DAVID database to interpret biological relevance. Results: Microarray analysis identified 51 differentially expressed miRNAs. Among these, 24 exhibited consistent expression patterns in high-marbling breeds compared to the low-marbling LIM breed. Bioinformatic analysis of the 4941 predicted target genes of these 24 miRNAs revealed significant enrichment in pathways crucial for marbling, including the adipocytokine, AMPK, MAPK, and PI3K-Akt signaling pathways, as well as biological processes such as cell differentiation and lipid homeostasis. Notably, miR-34a and miR-149-5p emerged as significant regulators, with miR-34a targeting genes like SIRT1, HMGA2, PTPN11, VEGFA, FGF1, FGF2, and BRAF, and miR-149-5p influencing adipogenesis and lipid metabolism through its association with crucial KEGG pathways such as PI3K–Akt, MAPK, PPAR, TGF-β, cAMP, and Wnt signaling, all of which collectively influence adipocyte differentiation, lipid metabolism, cell cycle control, and angiogenesis. Conclusions: The findings underscore identified miRNAs’ possible coordinated regulatory role, particularly miR-34a and miR-149-5p, in the complex molecular mechanisms governing IMF deposition in cattle, providing potential targets for improving beef quality. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

13 pages, 2207 KiB  
Article
Regulatory Role of lncRNA MEG3 Silencing on PI3K/GSK3β/Tau Pathway in a High-Glucose-Induced Cell Model
by Lütfiye Ozpak
Int. J. Mol. Sci. 2025, 26(16), 7944; https://doi.org/10.3390/ijms26167944 - 18 Aug 2025
Viewed by 67
Abstract
This study investigated the regulatory role of the long non-coding RNA maternally expressed gene 3 (MEG3) in tau hyperphosphorylation and insulin signaling (PI3K/AKT1/GSK3β) under high-glucose (HG)-induced neurotoxic conditions mimicking Alzheimer’s disease pathology. To explore the function of MEG3 within [...] Read more.
This study investigated the regulatory role of the long non-coding RNA maternally expressed gene 3 (MEG3) in tau hyperphosphorylation and insulin signaling (PI3K/AKT1/GSK3β) under high-glucose (HG)-induced neurotoxic conditions mimicking Alzheimer’s disease pathology. To explore the function of MEG3 within a hyperglycemic (Hyp) model, MEG3 was silenced using small interfering RNA (siRNA) assay, followed by Western blot analysis, qRT-PCR, and network analyses. The siMEG3 + Hyp group had lower levels of AKT1 (0.48-fold) and PI3K (0.52-fold) than did the Hyp group. In the siMEG3 + Hyp group, GSK3β (2.51-fold) and TNFα (2.38-fold) expressions were higher than were those in the Hyp group, while in the siMEG3 group, GSK3β (4.59-fold), microtubule-associated protein TAU (MAPT, TAU) (6.37-fold), interleukin (IL)1β (5.67-fold), IL6 (3.29-fold), and tumor necrosis factor-α (TNFα) (3.06-fold) were all significantly upregulated in comparison to the control group. A higher level of p-tau protein was seen in the siMEG3 group in comparison to the control group, as well as in the siMEG3 + Hyp group in comparison to the Hyp group. Gene ontology analysis following MEG3 administration showed that genes downstream of the PI3K pathway were suppressed, whereas genes regulating the neuroinflammatory response were upregulated. The results suggest that the lncRNA MEG3 may be a promising therapeutic target in HG-induced neurodegenerative AD. Full article
(This article belongs to the Special Issue Role of MicroRNAs in Human Diseases: 2nd Edition)
Show Figures

Figure 1

25 pages, 1499 KiB  
Systematic Review
Endothelial and Cardiovascular Effects of Naringin: A Systematic Review
by Jose A. Adams, Arkady Uryash, Alfredo Mijares, Jose Miguel Eltit and Jose R. Lopez
Nutrients 2025, 17(16), 2658; https://doi.org/10.3390/nu17162658 - 17 Aug 2025
Viewed by 239
Abstract
Background/Objectives: Naringin, a major flavonoid found in citrus fruits, has garnered significant attention over the past two decades for its potential cardiovascular benefits. This systematic review evaluates the effects of naringin on endothelial function and myocardial performance, with particular emphasis on ischemia-reperfusion (I/R) [...] Read more.
Background/Objectives: Naringin, a major flavonoid found in citrus fruits, has garnered significant attention over the past two decades for its potential cardiovascular benefits. This systematic review evaluates the effects of naringin on endothelial function and myocardial performance, with particular emphasis on ischemia-reperfusion (I/R) injury, based on the literature published from January 2000 to June 2025. Methods: The review was conducted in accordance with PRISMA 2020 guidelines. A comprehensive search of PubMed, Scopus, EMBASE, and Web of Science databases was performed using key terms including “naringin”, “cardiovascular”, “endothelial function”, “atherosclerosis”, and “ischemia-reperfusion.” A total of 62 studies were included and categorized into three domains: cellular models, animal studies, and human trials. Risk of bias assessments were conducted for each study type using appropriate tools. Results: Naringin consistently exhibited antioxidant, anti-inflammatory, and vasoprotective effects across all study types. Mechanistic studies highlighted the modulation of key signaling pathways, including PI3K/Akt, NF-κB, Nrf2, the renin-angiotensin system (RAS), and enhancement of KATP channel expression, as well as its ability to inhibit apoptosis, autophagy, and ferroptosis. In animal models, naringin improved endothelium-dependent vasorelaxation, reduced infarct size, and preserved myocardial function. Although limited, human trials reported beneficial effects on lipid profiles, arterial stiffness, and adiponectin levels. Conclusions: Naringin demonstrates strong potential as a dietary adjunct for cardiovascular protection, especially in the context of ischemic injury and vascular dysfunction. Further well-designed clinical trials are needed to define optimal dosing strategies and improve its bioavailability in humans. Full article
Show Figures

Figure 1

21 pages, 1395 KiB  
Article
Unlocking the Anti-Breast Cancer Potential of Aralia chinensis L.
by Juan Xue, Lei Li, Yongjia Shu, Chengshi Xie, Tian Lu and Huifang Chai
Curr. Issues Mol. Biol. 2025, 47(8), 662; https://doi.org/10.3390/cimb47080662 - 16 Aug 2025
Viewed by 149
Abstract
Aralia chinensis L. has shown potential in breast cancer treatment, yet its pharmacodynamically active components and mechanisms remain undefined. To systematically identify the bioactive constituents absorbed into the bloodstream and elucidate their multi-target mechanisms against breast cancer, we employed ultra-high-performance liquid chromatography in [...] Read more.
Aralia chinensis L. has shown potential in breast cancer treatment, yet its pharmacodynamically active components and mechanisms remain undefined. To systematically identify the bioactive constituents absorbed into the bloodstream and elucidate their multi-target mechanisms against breast cancer, we employed ultra-high-performance liquid chromatography in conjunction with Q Exactive Orbitrap mass spectrometry (UHPLC-Q Exactive Orbitrap-MS) alongside serum pharmacochemistry to analyze the chemical constituents of total saponins derived from A. chinensis (TSAC) and to identify the blood-absorbed prototypes in a rat model. Network pharmacology predicted targets and pathways of serum prototypes, validated by molecular docking and in vitro experiments. We identified 38 triterpenoid saponins, 3 steroidal saponins, and 8 triterpenoids in TSAC, with 22 prototype compounds detected in serum. An integrative analysis encompassing 486 compound targets and 1747 genes associated with breast cancer elucidated critical pathways, notably the PI3K-Akt signaling pathway and resistance mechanisms to EGFR tyrosine kinase inhibitors. Molecular docking confirmed strong binding of araloside A and elatoside L to SRC, PIK3R1, PIK3CA, STAT3, and EGFR. In MCF-7 cells, TSAC suppressed proliferation and migration while downregulating Src, PI3K, and EGFR expression at the gene and protein levels. This study successfully identified TSAC’s serum-absorbed bioactive components and demonstrated their anti-breast cancer effects via multi-target mechanisms involving the Src/PI3K/EGFR axis, providing a crucial pharmacological foundation for developing A. chinensis-derived breast cancer therapies. Full article
(This article belongs to the Special Issue Natural Compounds: An Adjuvant Strategy in Cancer Management)
Show Figures

Figure 1

34 pages, 3045 KiB  
Review
Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target
by Noah Brandl, Rebecca Seitz, Noah Sendtner, Martina Müller and Karsten Gülow
Antioxidants 2025, 14(8), 1002; https://doi.org/10.3390/antiox14081002 - 16 Aug 2025
Viewed by 324
Abstract
Reactive oxygen species (ROS) act as double-edged swords in cancer biology—facilitating tumor growth, survival, and metastasis at moderate levels while inducing oxidative damage and cell death when exceeding cellular buffering capacity. To survive under chronic oxidative stress, cancer cells rely on robust antioxidant [...] Read more.
Reactive oxygen species (ROS) act as double-edged swords in cancer biology—facilitating tumor growth, survival, and metastasis at moderate levels while inducing oxidative damage and cell death when exceeding cellular buffering capacity. To survive under chronic oxidative stress, cancer cells rely on robust antioxidant systems such as the glutathione (GSH) and thioredoxin (Trx), and superoxide dismutases (SODs). These systems maintain redox homeostasis and sustain ROS-sensitive signaling pathways including MAPK/ERK, PI3K/Akt/mTOR, NF-κB, STAT3, and HIF-1α. Targeting the antioxidant defense mechanisms of cancer cells has emerged as a promising therapeutic strategy. Inhibiting the glutathione system induces ferroptosis, a non-apoptotic form of cell death driven by lipid peroxidation, with compounds like withaferin A and altretamine showing strong preclinical activity. Disruption of the Trx system by agents such as PX-12 and dimethyl fumarate (DMF) impairs redox-sensitive survival signaling. Trx reductase inhibition by auranofin or mitomycin C further destabilizes redox balance, promoting mitochondrial dysfunction and apoptosis. SOD1 inhibitors, including ATN-224 and disulfiram, selectively enhance oxidative stress in tumor cells and are currently being tested in clinical trials. Mounting preclinical and clinical evidence supports redox modulation as a cancer-selective vulnerability. Pharmacologically tipping the redox balance beyond the threshold of cellular tolerance offers a rational and potentially powerful approach to eliminate malignant cells while sparing healthy tissue, highlighting novel strategies for targeted cancer therapy at the interface of redox biology and oncology. Full article
Show Figures

Figure 1

Back to TopTop