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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,143)

Search Parameters:
Keywords = HIF1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 3565 KB  
Article
Downregulation of miR-27a-3p Modulates TGF-β Signaling and Dysregulates Metabolism in Glioblastoma
by Augusto Ferreira Weber, Juliete Nathali Scholl, Camila Kehl Dias, Vinícius Pierdoná Lima, Tamires de Bona, Renata Marschner, Arieli Cruz de Sousa, Fábio Klamt and Fabrício Figueiró
Int. J. Mol. Sci. 2025, 26(17), 8729; https://doi.org/10.3390/ijms26178729 (registering DOI) - 8 Sep 2025
Abstract
Several microRNAs (miRNAs) are key influencers of tumor microenvironment (TME) cell plasticity, regulating the progression of various tumor types such as glioblastoma (GBM). Differential expressions of miR-27a-3p and miR-155-5p in GBM cells and biopsies have already been described as markers of tumor subtype [...] Read more.
Several microRNAs (miRNAs) are key influencers of tumor microenvironment (TME) cell plasticity, regulating the progression of various tumor types such as glioblastoma (GBM). Differential expressions of miR-27a-3p and miR-155-5p in GBM cells and biopsies have already been described as markers of tumor subtype and progression. We aimed to evaluate the cellular and molecular impacts of inhibiting these two overexpressed miRNAs in GBM cell lines. A172 cells were transfected with miR-27a-3p and miR-155-5p inhibitors, and the effects on cellular processes and the expression of malignancy-related genes were analyzed by flow cytometry and qPCR, respectively. Thus, several cellular characteristics in A172 cells were modulated; however, only the inhibition of miR-27a-3p resulted in apoptosis, reduced glucose uptake, and a decrease in mitochondrial membrane potential. Both inhibitors modulated metabolic and immunological targets, negatively regulating genes in the glycolysis pathway and modulating other metabolic pathways involving glutamine and fatty acids, for example. Additionally, it modulates the TGF-β pathway, which can influence the GBM microenvironment due to its immunosuppressive role in advanced tumors. miR-27a-3p appears to be a pivotal factor in the functional duality of TGF-β and its interaction with HIF1A in the hypoxic tumor environment, modulating SMAD partners or TGF-β pathway inhibitors. Here, we demonstrate the importance of inhibiting overexpressed miRNAs, particularly miR-27a-3p, in modulating key pathways for tumor cell survival. The results of this work provide new insights into potential targets for immune-metabolic interactions in the TME and their implications for tumorigenesis, shedding light on new therapeutic approaches for GBM. Full article
Show Figures

Graphical abstract

17 pages, 1707 KB  
Article
Combined Hesperidin and Gemcitabine Therapy Modulates Apoptosis and Angiogenesis Pathways in ISHIKAWA Human Endometrial Adenocarcinoma Cells
by Yasemin Afşin, İlhan Özdemir, Veysel Toprak, Mehmet Cudi Tuncer and Şamil Öztürk
Medicina 2025, 61(9), 1599; https://doi.org/10.3390/medicina61091599 - 4 Sep 2025
Viewed by 316
Abstract
Background and Objectives: Endometrial adenocarcinoma is among the most prevalent malignancies of the female reproductive system, and therapeutic options remain limited, particularly in advanced stages. In recent years, natural agents, especially flavonoids, have gained considerable interest for their capacity to enhance the effectiveness [...] Read more.
Background and Objectives: Endometrial adenocarcinoma is among the most prevalent malignancies of the female reproductive system, and therapeutic options remain limited, particularly in advanced stages. In recent years, natural agents, especially flavonoids, have gained considerable interest for their capacity to enhance the effectiveness of chemotherapeutic drugs and modulate tumor-related molecular mechanisms. Hesperidin, a citrus-derived flavonoid, is recognized for its antioxidant and anti-inflammatory effects, while Gemcitabine, a nucleoside analog, is widely used in cancer treatment. Investigating their combined effects on endometrial carcinoma cells could yield novel insights into multimodal therapeutic development. This current study aimed to assess the impact of Hesperidin (Hes) and Gemcitabine (Gem) on ISHIKAWA cells, a human endometrial adenocarcinoma model, with particular attention to pathways associated with hypoxia, angiogenesis, apoptosis, and oxidative stress. Materials and Methods: ISHIKAWA cells were treated with varying concentrations of Hes (50–200 µM) and Gem (10–50 nM), either individually or together, for 24 and 48 h. Cell viability was determined using the MTT assay, while apoptosis was measured by Caspase-3/7 activity and NucBlue nuclear staining. Intracellular reactive oxygen species (ROS) generation was quantified via DCFH-DA fluorescence. Expression levels of HIF-1α, VEGF, Bax, Bcl-2, and Caspase-3 were examined by RT-qPCR. Synergistic interactions were analyzed with the Chou–Talalay combination index. Biological enrichment was further explored using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Results: Both Hes and Gem significantly decreased ISHIKAWA cell viability in a concentration- and time-dependent manner (p < 0.001). The combined treatment induced stronger apoptotic effects, as reflected by increased Caspase-3/7 activity and nuclear morphological changes. RT-qPCR demonstrated upregulation of Bax and Caspase-3, together with downregulation of Bcl-2, HIF-1α, and VEGF. While Hes reduced intracellular ROS, Gem elevated it; their combination produced a balanced oxidative response. All dose combinations displayed strong synergism (CI < 1). GO and KEGG enrichment confirmed the involvement of apoptosis-, angiogenesis-, and hypoxia-related pathways. Conclusions: Co-treatment with Hes and Gem exhibits synergistic anticancer activity in endometrial cancer cells by promoting apoptosis, suppressing angiogenesis- and hypoxia-related gene expression, and modulating oxidative stress. This combined therapeutic approach highlights its potential as a promising adjuvant option, warranting further evaluation in in vivo and translational studies. Full article
(This article belongs to the Special Issue Gynecological and Oncological Diseases in the Aged)
Show Figures

Figure 1

26 pages, 10956 KB  
Article
Investigation of Anticancer Properties of Newly Synthesized Pyridazine-Based Inhibitors in Mouse and Human Breast Cancer Cell Line
by Kübra Acikalin Coskun, Elif Cansu Abay, Mehmet Gumus, Ayşe Büşranur Çelik, Levent Gulum, Irfan Koca and Yusuf Tutar
Biology 2025, 14(9), 1193; https://doi.org/10.3390/biology14091193 - 4 Sep 2025
Viewed by 230
Abstract
Background: Breast cancer is the most common cancer among women. Although doxorubicin (DOX) is widely used in its treatment, its dose-dependent toxicity and the development of drug resistance reduce its therapeutic efficacy. Therefore, this study aims to identify a novel anticancer agent that [...] Read more.
Background: Breast cancer is the most common cancer among women. Although doxorubicin (DOX) is widely used in its treatment, its dose-dependent toxicity and the development of drug resistance reduce its therapeutic efficacy. Therefore, this study aims to identify a novel anticancer agent that is more effective than DOX, inhibits cancer cell growth, and is less toxic to healthy cells. Methods: The cytotoxic effects of DOX and 2S-series molecules were evaluated on human (MDA-MB-231) and mouse (4T1) TNBC breast cancer cell lines and healthy breast epithelial (hTERT) cells using MTT assays at 48 and 72 h to screen functional similarities and possible differences upon drug/inhibitor treatment. Apoptosis and cell cycle analysis were analyzed by flow cytometry. Gene expression profiles were assessed by qPCR, and binding interactions with Hsp90 were examined via molecular docking. Results: 2S-5 exhibited IC50 values of 6.21 µM (MDA-MB-231) and 7.04 µM (4T1), while 2S-13 showed IC50 values of 7.73 µM and 8.21 µM, respectively. Both compounds demonstrated selective cytotoxicity against cancer cells. Gene expression and pathway analysis revealed that 2S-13 modulated the PI3K-Akt, MAPK, apoptosis, and HIF-1 pathways, showing broader modulation than DOX. Conclusions: 2S-13 appears to be a promising drug candidate, particularly in the MDA-MB-231 cell line. However, the current findings are limited to in vitro models. Further in vivo studies and pharmacokinetic analyses are required to validate its therapeutic potential, assess long-term efficacy and safety, and explore its resistance profile and molecular mechanisms in more detail. Full article
(This article belongs to the Special Issue Advances in Biological Breast Cancer Research (2nd Edition))
Show Figures

Figure 1

18 pages, 4672 KB  
Article
Environmental Hazards and Chemoresistance in OTSCC: Molecular Docking and Prediction of Paclitaxel and Imatinib as BCL2 and EGFR Inhibitors
by Nishant Kumar Singh, Prankur Awasthi, Agrika Gupta, Nidhi Anand, Balendu Shekher Giri and Saba Hasan
Biology 2025, 14(9), 1174; https://doi.org/10.3390/biology14091174 - 2 Sep 2025
Viewed by 384
Abstract
Oral tongue squamous cell carcinoma (OTSCC) is a common type of oral cancer influenced by genetic, epigenetic, and environmental factors like exposure to environmental toxins. These environmental toxins can decrease the effectiveness of established chemotherapy drugs, such as Irinotecan, used in OTSCC treatment. [...] Read more.
Oral tongue squamous cell carcinoma (OTSCC) is a common type of oral cancer influenced by genetic, epigenetic, and environmental factors like exposure to environmental toxins. These environmental toxins can decrease the effectiveness of established chemotherapy drugs, such as Irinotecan, used in OTSCC treatment. Bioinformatics, drug discovery, and machine learning techniques were employed to investigate the impact of Irinotecan on OTSCC patients by identifying targets and signaling pathways, including those that positively influence protein phosphorylation, protein tyrosine kinase activity, the PI3K-Akt (Phosphatidylinositol 3-kinase- Protein Kinase B) signaling system, cancer pathways, focal adhesion, and the HIF-1 (Hypoxia-Inducible Factor 1) signaling pathway. Later, the protein–protein interactions (PPIs) network, along with twelve cytoHubba approaches to finding the most interacting molecule, was employed to find the important proteins BCL2 and EGFR. Drugs related to BCL2 and EGFR were extracted from the DGIdb database for further molecular docking. Molecular docking revealed that Docetaxel, Paclitaxel, Imatinib, Ponatinib, Ibrutinib, Sorafenib, and Etoposide showed more binding affinity than Irinotecan (i.e., −9.8, −9.6). Of these, Paclitaxel (−10.3, −11.4) and Imatinib (−9.9, −10.4) are common in targeting BCL2 and EGFR. Using these identified candidate genes and pathways, we may be able to uncover new therapeutic targets for the treatment of OTSCC. Furthermore, molecular dynamics (MD) simulations were performed for selected ligand–receptor complexes, revealing stable binding interactions and favorable energetic profiles that supported the docking results and strengthened the reliability of the proposed drug repurposing strategy. Full article
(This article belongs to the Special Issue Head and Neck Cancer: Current Advances and Future Perspectives)
Show Figures

Graphical abstract

16 pages, 2669 KB  
Review
Latest Nanoparticles to Modulate Hypoxic Microenvironment in Photodynamic Therapy of Cervical Cancer: A Review of In Vivo Studies
by Dorota Bartusik-Aebisher, Mohammad A. Saad, Agnieszka Przygórzewska, Paweł Woźnicki and David Aebisher
Int. J. Mol. Sci. 2025, 26(17), 8503; https://doi.org/10.3390/ijms26178503 - 1 Sep 2025
Viewed by 303
Abstract
Photodynamic therapy (PDT) is a promising, minimally invasive treatment for cervical cancer, but its efficacy is significantly limited by hypoxia—oxygen deficiency in the tumour microenvironment. The aim of this study was to present strategies to counteract hypoxia in PDT using the latest nanotechnologies. [...] Read more.
Photodynamic therapy (PDT) is a promising, minimally invasive treatment for cervical cancer, but its efficacy is significantly limited by hypoxia—oxygen deficiency in the tumour microenvironment. The aim of this study was to present strategies to counteract hypoxia in PDT using the latest nanotechnologies. Based on a review of the literature available in PubMed/MEDLINE, Scopus, and Web of Science databases, covering the period from January 2024 to March 2025, nine original in vivo studies were identified that investigated the use of nanoparticle-based strategies to overcome hypoxia and enhance the efficacy of PDT in cervical cancer. A variety of approaches to improve tumour oxygenation are described, including the catalytic decomposition of hydrogen peroxide (H2O2) with manganese oxide (MnO2), the use of bimetallic nanozymes (e.g., Au2Pt), and FeOOH structures and oxygen storage and control systems (e.g., endoperoxides). Strategies to reduce oxygen consumption by cancer cells, such as nitric oxide (NO) release or inhibition of mitochondrial oxidative phosphorylation, are also discussed. The review shows that appropriately designed nanoparticles can effectively counteract hypoxia, enhancing the efficacy of PDT by intensifying reactive oxygen species (ROS) generation and modulating HIF-1α factor expression. The strategies presented here have the potential to significantly improve the efficacy of photodynamic therapy in the treatment of cervical cancer, especially under conditions of limited oxygen availability. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Figure 1

23 pages, 5430 KB  
Article
β-Caryophyllene Ameliorates Thioacetamide-Induced Liver Fibrosis in Rats: A Preventative Approach
by Lujain Bader Eddin, Amar Mahgoub, Saeeda Almarzooqi, Ernest Adeghate, Sandeep B. Subramanya and Shreesh Ojha
Int. J. Mol. Sci. 2025, 26(17), 8493; https://doi.org/10.3390/ijms26178493 - 1 Sep 2025
Viewed by 236
Abstract
Liver fibrosis is associated with increased rates of morbidity and mortality. At present, there are no specific treatments that can directly reverse hepatic fibrosis. The endocannabinoid system has been found to play a significant role in regulating the development and progression of liver [...] Read more.
Liver fibrosis is associated with increased rates of morbidity and mortality. At present, there are no specific treatments that can directly reverse hepatic fibrosis. The endocannabinoid system has been found to play a significant role in regulating the development and progression of liver diseases, in addition to having protective effects. In this study, we investigate the protective potential of β-Caryophyllene (BCP) against Thioacetamide (TAA)-induced liver fibrosis. Wistar rats were injected with TAA (200 mg/kg) three times per week for 8 weeks to induce liver fibrosis. They also received oral BCP before the TAA injections. AM630 (1 mg/kg) was administered to confirm the CB2 receptor-dependent effect of BCP. The BCP treatment (50 mg/kg) protected against cell injury and potentiated antioxidant defense by replenishing hepatic GSH, improving catalase activity, and inhibiting the formation of MDA. The co-administration of BCP mitigated the TAA-induced inflammatory response by decreasing the release of proinflammatory cytokines. Histological examination showed preserved cellular integrity, decreased collagen deposits with other extracellular matrix proteins, and low levels of myofibroblast activation. In addition, the BCP-treated rats demonstrated upregulated sirtuin 1 (SIRT1) expression, which had a direct inhibitory effect on hypoxia inducible factor (HIF-1α). AM630 pre-treatment inhibited all the aforementioned protective mechanisms of BCP. Based on our findings, BCP exerts protective effects in liver fibrosis, which can be attributed to its agonist action on CB2 receptors. This study provides preclinical evidence of the potential preventative benefits of BCP in liver fibrosis. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Graphical abstract

52 pages, 10321 KB  
Article
Prognostic Significance of WWOX/HIF1A Ratio in Cancer Subtypes: Insights into Metabolism, ECM, and EMT
by Izabela Baryła, Raneem Y. Hammouz, Kinga Maciejek and Andrzej K. Bednarek
Biology 2025, 14(9), 1151; https://doi.org/10.3390/biology14091151 - 1 Sep 2025
Viewed by 341
Abstract
WWOX and HIF1α proteins are involved in cancer progression; their functions are closely related. WWOX binds HIF1α through its WW domains, sequestering it in the cytoplasm and inhibiting its transcriptional activity. This study evaluates the prognostic significance of the WWOX/HIF1A interaction [...] Read more.
WWOX and HIF1α proteins are involved in cancer progression; their functions are closely related. WWOX binds HIF1α through its WW domains, sequestering it in the cytoplasm and inhibiting its transcriptional activity. This study evaluates the prognostic significance of the WWOX/HIF1A interaction across cancers, breast cancer subtypes, glioblastoma (GBM), low-grade glioma (LGG), and hepatocellular carcinoma (HCC) through gene expression and pathway analysis focused on metabolism, ECM, and epithelial–mesenchymal transition. In breast cancer, metabolic pathways correlated with good prognosis in basal subtypes. HER2 subtypes showed enrichment in DNA replication pathways. Luminal A subtypes showed favourable prognosis via TNF and PI3K/AKT signalling, while luminal B subtypes had poor prognosis tied to metabolic activity; genes associated with good prognosis mirrored those tied to poor prognosis in luminal A. In HCC, enhanced metabolic activity was associated with good prognosis. In contrast, poor prognosis involved TNF signalling and cytoskeleton-related pathways, indicating more aggressive tumour behaviour. In LGG, good prognosis was linked to metabolic and cAMP pathways, while poor outcomes involved TNF, cell cycle, apoptosis, and focal adhesion pathways. GBM showed similar patterns: metabolic and cAMP pathways indicated better outcomes, while NFKB, TNF, JAK-STAT, and PI3K/AKT pathways marked poor prognosis. These findings suggest the WWOX/HIF1A ratio is a robust prognostic marker and a possible guide for developing targeted treatments. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

19 pages, 1548 KB  
Review
Oxygen-Mediated Molecular Mechanisms Involved in Intestinal Ischemia and Reperfusion Injury
by Paraschos Archontakis-Barakakis, Theodoros Mavridis and Athanasios Chalkias
Int. J. Mol. Sci. 2025, 26(17), 8398; https://doi.org/10.3390/ijms26178398 - 29 Aug 2025
Viewed by 322
Abstract
The gastrointestinal tract is affected by multiple ailments that manifest with similar chemical, subcellular, and cellular changes, such as those in intestinal ischemia–reperfusion injury (IRI). The main chemical changes that are described under IRI conditions include the depletion of oxygen available for normal [...] Read more.
The gastrointestinal tract is affected by multiple ailments that manifest with similar chemical, subcellular, and cellular changes, such as those in intestinal ischemia–reperfusion injury (IRI). The main chemical changes that are described under IRI conditions include the depletion of oxygen available for normal metabolism and the abundant production and increase in intracellular and extracellular concentrations of hydrogen peroxide and other reactive oxygen species (ROS). The enzymes causing this accumulation are xanthine dehydrogenase turning into xanthine oxidase, nicotinamide adenine dinucleotide phosphate oxidase, and nitric oxide synthase. The cellular changes revolve around an oxygen-sensing system that is responsive to varying oxygen levels, which has Hypoxia-Inducible Factors (HIFs) at its base. HIFs are transcription factors, the intracellular concentrations of which significantly increase under hypoxic conditions. Upon activation, they alter the expression of gene sets to ensure appropriate cellular adjustment to the hypoxic and IRI environment. Despite the primary regulation of the system involving oxygen, it is interconnected with multiple other subcellular and cellular functions. Thus, it represents a linchpin control mechanism of cellular adaptation. The effect of HIF activation in intestinal cells aims at preserving the structural integrity of the intestinal lining. The effect in different subtypes of leucocytes aims at immune system activation to protect against previously luminally located and subsequently invading pathogens and toxins. All in all, the HIF system is an integral part of cellular and tissue compensation against intestinal IRI. Full article
(This article belongs to the Special Issue New Molecular Insights into Ischemia/Reperfusion: 2nd Edition)
Show Figures

Figure 1

20 pages, 7746 KB  
Article
Silybin Mitigates Post-Myocardial Infarction Heart Failure in Mice via Modulation of HIF-1α-Driven Glycolysis and Energy Metabolism
by Mengyuan Wang, Jinhong Chen, Zhongzheng Zhang, Tianyu Wang, Jiaqi Zhao, Xiao Wang, Junyan Wang and Haowen Zhuang
Nutrients 2025, 17(17), 2800; https://doi.org/10.3390/nu17172800 - 28 Aug 2025
Viewed by 558
Abstract
Background: Post-myocardial infarction (MI) heart failure (HF) is characterized by myocardial energy metabolism disorder, with excessive glycolysis playing a key role in its progression. Silybin (SIL), a flavonoid derived from Silybum marianum, has demonstrated hepatoprotective and metabolic regulatory effects. However, the role of [...] Read more.
Background: Post-myocardial infarction (MI) heart failure (HF) is characterized by myocardial energy metabolism disorder, with excessive glycolysis playing a key role in its progression. Silybin (SIL), a flavonoid derived from Silybum marianum, has demonstrated hepatoprotective and metabolic regulatory effects. However, the role of this flavonoid in ameliorating post-myocardial infarction heart failure (post-MI HF) by modulating energy metabolism remains unclear. Methods: This study employed an oxygen–glucose deprivation (OGD) model to induce myocardial cell injury in vitro, with YC-1 treatment used to inhibit hypoxia-inducible factor-1α (HIF-1α) for mechanistic validation. A myocardial infarction-induced HF mouse model was used for in vivo experiments. Results: In vitro, SIL enhanced cell viability, increased ATP levels, and decreased lactate production and reactive oxygen species (ROS) accumulation in OGD-treated myocardial cells. SIL downregulated the mRNA and protein expression of HIF-1α, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), glucose transporter 1 (GLUT1), and lactate dehydrogenase A (LDHA) while inhibiting HIF-1α nuclear translocation. Furthermore, SIL suppressed glycolytic proteins (PFKFB3, GLUT1, and LDHA) in a manner comparable to the HIF-1α inhibitor YC-1. This confirms that SIL’s inhibition of glycolysis is HIF-1α-dependent. In vivo, SIL treatment improved cardiac function parameters (LVEF and LVFS) and attenuated left ventricular remodeling (LVID;d and LVID;s) in post-MI HF mice. Additionally, myocardial fibrosis markers were significantly reduced, accompanied by a decrease in the myocardial mRNA and protein expression of glycolytic proteins, including HIF-1α, PFKFB3, GLUT1, and LDHA. Conclusions: Silybin effectively ameliorates post-myocardial infarction heart failure through the HIF-1α-mediated regulation of glycolysis, leading to improved myocardial energy metabolism and enhanced cardiac function. Full article
Show Figures

Figure 1

18 pages, 1463 KB  
Article
Hypoxia-Driven Extracellular Vesicles Promote Pro-Metastatic Signalling in LNCaP Cells via Wnt and EMT Pathways
by Melissa Santos, Khansa Bukhari, Irem Peker-Eyüboğlu, Igor Kraev, Dafydd Alwyn Dart, Sigrun Lange and Pinar Uysal-Onganer
Biology 2025, 14(9), 1135; https://doi.org/10.3390/biology14091135 - 27 Aug 2025
Viewed by 507
Abstract
Prostate cancer (PCa) progression is shaped by the tumour microenvironment, where hypoxia promotes aggressiveness and contributes to therapy resistance. Extracellular vesicles (EVs), secreted under hypoxia, can deliver modified bioactive cargo that reprograms recipient cells. This study examined whether EVs from hypoxia-conditioned metastatic PCa [...] Read more.
Prostate cancer (PCa) progression is shaped by the tumour microenvironment, where hypoxia promotes aggressiveness and contributes to therapy resistance. Extracellular vesicles (EVs), secreted under hypoxia, can deliver modified bioactive cargo that reprograms recipient cells. This study examined whether EVs from hypoxia-conditioned metastatic PCa cells enhance malignant traits in cancerous and non-tumorigenic prostate cell lines via Wnt signalling and epithelial–mesenchymal transition (EMT). EVs from PC3 cells cultured under hypoxia (1% O2) or normoxia (21% O2) as control were applied to LNCaP (low metastatic potential) and PNT2 (non-tumorigenic) cells. PC3 hypoxia-derived EVs increased HIF-1α, upregulated mesenchymal markers (Vimentin, N-cadherin) and Wnt-related genes (Wnt3A, Wnt5A, Fzd7), and suppressed the epithelial marker E-cadherin. Functional assessment showed that LNCaP cells treated with PC3 hypoxia EVs showed greater motility and invasiveness, and PNT2 cells displayed transcriptomic reprogramming. These findings show that hypoxia-driven EVs can propagate pro-metastatic signalling in less aggressive and normal prostate cells. The findings highlight EVs as a potential therapeutic target in PCa progression. Full article
(This article belongs to the Special Issue Signalling Pathways in Cancer and Disease)
Show Figures

Graphical abstract

23 pages, 6781 KB  
Article
Characteristics of Polyphenols of Black Hulless Barley Bran and Its Anti-Diabetic Activity
by Junlin Deng, Tinghui Liu, Chen Xia, Litao Tong, Chunmei Gu, Zhiqiang Shi, Yuehang Yang, Ruiling Zhan, Zhuoya Xiang, Jian Chen, Yan Wan and Manyou Yu
Foods 2025, 14(17), 2994; https://doi.org/10.3390/foods14172994 - 27 Aug 2025
Viewed by 423
Abstract
Polyphenols play a crucial role in promoting human health. This study aims to investigate the polyphenols of black hulless barley bran (HBP) and evaluate their anti-diabetic mechanisms in vivo. Using UPLC-QTOF-MS/MS, 27 compounds were identified in HBP, including four phenolic acids, 14 flavonoids, [...] Read more.
Polyphenols play a crucial role in promoting human health. This study aims to investigate the polyphenols of black hulless barley bran (HBP) and evaluate their anti-diabetic mechanisms in vivo. Using UPLC-QTOF-MS/MS, 27 compounds were identified in HBP, including four phenolic acids, 14 flavonoids, and nine anthocyanidins. High contents of Chrysoeriol 7-O-glucuronide (42.09 mg/g), Cyanidin 3-O-glucoside (21.02 mg/g), and Cyanidin 3-O-(6″-O-malonyl)-glucoside (24.45 mg/g) were quantified via UPLC in HBP. Administration of HBP significantly reduced fasting blood glucose (FBG), improved glucose intolerance and lipid profiles, and alleviated liver and pancreatic damage in type 2 diabetic (T2DM) mice. Furthermore, it enhanced serum antioxidant enzyme activities and modulated inflammatory cytokines. Transcriptomic analysis revealed that HBP influenced signal transduction and the immune system, particularly in key signaling pathways, including Hippo, TGF-beta, HIF-1, and p53, associated with T2DM. Although HBP had minimal impact on gut microbiota diversity and SCFA levels, it presents a promising candidate for T2DM intervention through its multifaceted mechanisms. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Graphical abstract

15 pages, 1676 KB  
Article
Mitigating Oxidative Stress and Anti-Angiogenic State in an In Vitro Model of Preeclampsia by HY-12, an Organofluorine Hydrazone Antioxidant
by Zsuzsanna K. Zsengellér, Maxim Mastyugin, Adrianna R. Fusco, Bernadett Vlocskó, Maximilian Costa, Coryn Ferguson, Diana Pintye, Réka Eszter Sziva, Saira Salahuddin, Brett C. Young, Marianna Török and Béla Török
Curr. Issues Mol. Biol. 2025, 47(9), 680; https://doi.org/10.3390/cimb47090680 - 24 Aug 2025
Viewed by 672
Abstract
Preeclampsia (PE) is a hypertensive disorder impacting 5–7% of pregnancies globally. With no causative treatment available, diagnosed patients have limited therapeutic options, putting them at risk for pregnancy complications. The induction of oxidative stress by ROS—one of the major contributors in PE pathogenesis—causes [...] Read more.
Preeclampsia (PE) is a hypertensive disorder impacting 5–7% of pregnancies globally. With no causative treatment available, diagnosed patients have limited therapeutic options, putting them at risk for pregnancy complications. The induction of oxidative stress by ROS—one of the major contributors in PE pathogenesis—causes downstream signaling and production of anti-angiogenic factors, such as sFLT1 and sEng. The anti-angiogenic factors may cause endothelial and trophoblast dysfunction, contributing to the development of hypertension, proteinuria, and in severe cases, eclampsia. To target placental oxidative stress, we developed and evaluated an organofluorine hydrazone antioxidant, HY-12, in vitro. Human trophoblast (HTR8/SVneo) cells were incubated with hydrogen peroxide to induce oxidative stress and act as a model of PE. The goal of the study was to assess the efficacy of HY-12 and its ability to reduce cell injury, mitochondrial stress, and anti-angiogenic response. In our human trophoblast-based assays, pre-treatment with HY-12 reduced mitochondrial-derived ROS production in cells exposed to hydrogen peroxide, proving its ability to alleviate the oxidative stress associated with the pathogenesis of PE. HY-12 reduced HIF1A expression and sFLT1 protein expression in H2O2-exposed HTR8 cells. Furthermore, HY-12 improved the activity of the mitochondrial electron chain enzyme cytochrome C oxidase (COX) in the hydrogen-peroxide-treated HTR8/SVneo cells, which is a promising attribute of the compound. In reducing placental trophoblast oxidative stress, HY-12 shows promise as a potential treatment of preeclampsia. In vivo studies are warranted to further determine the efficacy of this compound. Full article
Show Figures

Graphical abstract

14 pages, 2103 KB  
Article
Comparative Analysis of Gene and MicroRNA Expression in Subcutaneous Adipose Tissue in Metabolically Healthy and Unhealthy Obesity
by Natalia O. Markina, Georgy A. Matveev, Ksenia A. Zasypkina, Natalia V. Khromova, Alina Yu. Babenko and Evgeny V. Shlyakhto
Int. J. Mol. Sci. 2025, 26(17), 8212; https://doi.org/10.3390/ijms26178212 - 24 Aug 2025
Viewed by 452
Abstract
Metabolically healthy (MHO) and unhealthy obesity (MUO) exhibit distinct molecular genetic mechanisms underlying metabolic disorders. Studying gene and microRNA expression in subcutaneous adipose tissue (SAT) may reveal key pathogenetic differences between these phenotypes. We compared the expression of genes (ADIPOQ, HIF1A, CCL2) and [...] Read more.
Metabolically healthy (MHO) and unhealthy obesity (MUO) exhibit distinct molecular genetic mechanisms underlying metabolic disorders. Studying gene and microRNA expression in subcutaneous adipose tissue (SAT) may reveal key pathogenetic differences between these phenotypes. We compared the expression of genes (ADIPOQ, HIF1A, CCL2) and microRNAs (miR-142-3p, miR-155, miR-378) in SAT between MHO and MUO patients and assessed their association with metabolic parameters. The study included 39 obese patients (19 MHO, 20 MUO) and 10 healthy controls. SAT biopsies were analyzed using real-time PCR. Correlations with clinical and metabolic markers were evaluated. Obese patients showed decreased ADIPOQ (p = 0.039) and miR-142 (p = 0.008) expression and increased CCL2 (p = 0.004), miR-155 (p = 0.017), and miR-378 (p = 0.04) expression compared to the controls. MUO patients exhibited higher HIF1A expression (p = 0.03) and strong correlations between CCL2 and dyslipidemia (total cholesterol, triglycerides)/dysglycemia (fasting glucose) (r = 0.45, p = 0.03; r = 0.52, p = 0.01; r = 0.63, p = 0.001, respectively). miR-142 negatively correlated with fibrosis markers, while miR-378 was linked to insulin resistance. The differential gene and microRNA expression highlights the role of inflammation, hypoxia, and fibrosis in MUO pathogenesis. miR-142-3p, miR-155, and miR-378 may serve as potential biomarkers for metabolic risk stratification and therapeutic targets. Full article
Show Figures

Figure 1

18 pages, 886 KB  
Review
Chinese Medicine-Derived Natural Compounds and Intestinal Regeneration: Mechanisms and Experimental Evidence
by Fengbiao Guo and Shaoyi Zhang
Biomolecules 2025, 15(9), 1212; https://doi.org/10.3390/biom15091212 - 22 Aug 2025
Viewed by 546
Abstract
Intestinal regeneration is essential for maintaining epithelial integrity and repairing mucosal damage caused by inflammation, infections, or injuries. Traditional Chinese Medicine (TCM) has long utilized herbal remedies for gastrointestinal disorders, and accumulating evidence highlights that natural compounds derived from TCM possess significant regenerative [...] Read more.
Intestinal regeneration is essential for maintaining epithelial integrity and repairing mucosal damage caused by inflammation, infections, or injuries. Traditional Chinese Medicine (TCM) has long utilized herbal remedies for gastrointestinal disorders, and accumulating evidence highlights that natural compounds derived from TCM possess significant regenerative potential. This review summarizes the multifaceted mechanisms by which these bioactive compounds promote intestinal healing. Key actions include the stimulation of intestinal stem cell (ISC) proliferation and differentiation, the modulation of inflammatory responses, the reinforcement of epithelial barrier integrity, the attenuation of oxidative stress, and the reshaping of the gut microbiota. Representative compounds such as Astragalus polysaccharides, berberine, curcumin, puerarin, and flavonoids like quercetin exhibit these effects through signaling pathways, including HIF-1, Wnt/β-catenin, NF-κB, Nrf2, and IL-22. Evidence from in vitro organoid models and in vivo studies in colitis, radiation injury, antibiotic-associated diarrhea, and intestinal dysmotility and diarrhea models demonstrates that these compounds enhance crypt villus regeneration, preserve tight junctions, and improve clinical outcomes. The holistic, multi-target actions of Chinese medicine-derived natural products make them promising candidates for therapeutic strategies aimed at intestinal repair. Further clinical validation and mechanistic studies are warranted to facilitate their integration into modern gastrointestinal medicine. Full article
(This article belongs to the Special Issue Natural Bioactives as Leading Molecules for Drug Development)
Show Figures

Figure 1

22 pages, 11866 KB  
Article
Study on the Mechanism of RuHaoDaShi Granules in Treating H1N1 Viral Pneumonia Based on Network Pharmacology and Experimental Validation
by Aixin Chen, Tianhang Chen, Yu He, Jiehong Yang and Haitong Wan
Pathogens 2025, 14(8), 834; https://doi.org/10.3390/pathogens14080834 - 21 Aug 2025
Viewed by 484
Abstract
Objective: This study aims to investigate the pharmacodynamic effects and underlying mechanisms of the Chinese herbal formula RuHaoDaShi (RHDS) granules against the influenza virus in experimental models. Methods: This study aims to employ network pharmacology to identify the active components of RHDS and [...] Read more.
Objective: This study aims to investigate the pharmacodynamic effects and underlying mechanisms of the Chinese herbal formula RuHaoDaShi (RHDS) granules against the influenza virus in experimental models. Methods: This study aims to employ network pharmacology to identify the active components of RHDS and its potential targets and mechanisms of action against H1N1. The molecular docking approach validated the interactions between the core targets and the RHDS compounds. In vitro, the antiviral activity of RHDS was assessed by therapeutic, prophylactic, and premixed administration to H1N1-infected A549 cells. An in vivo experiment was conducted using a mouse H1N1 pneumonia model. The model was treated with a dose of 1.04, 2.08, and 4.16 g/kg of RHDS, administered via gavage daily. The study’s objective was to evaluate the antiviral activity and mechanism of action of RHDS in mice. Mice were evaluated on day 6 by assessing survival, viral load (RT-qPCR), lung pathology (HE staining), inflammatory cytokines (ELISA, immunohistochemistry), and ferroptosis markers (WB, qPCR). Results: Network pharmacology identified 77 biologically active RHDS compounds (e.g., quercetin and kaempferol) and 32 core targets common to RHDS, H1N1, and ferroptosis. Molecular docking was used to verify a high affinity for binding between the core targets HIF-1α, MAPK3, and key RHDS compounds. In vitro studies demonstrated that RHDS exhibited protective properties against H1N1-infected cells, with the therapeutic delivery method proving the most efficacious. In vivo studies have shown that RHDS reduces mortality, lung index, and viral load in mice while attenuating histopathological damage. The study demonstrated a reduction in the release of inflammatory cytokines, including IL-6, IFN-γ, and IL-17A, and decreased expression levels of MPO and F4/80 proteins in lung tissue. Mechanistically, the administration of RHDS resulted in the up-regulation of the expression levels of GPX4, SLC7A11, and Nrf2 proteins while concomitantly inhibiting the expression of HIF-1α, COX2, and ACSL4. These findings confirm the modulatory effect of RHDS on the GPX4/SLC7A11/Nrf2 pathway. Conclusions: RHDS demonstrated a protective effect against H1N1-induced cytopathy in vitro and was effective in attenuating H1N1-induced pneumonia in murine models. The study suggests that RHDS has antiviral potential to treat H1N1 viral pneumonia by modulating inflammatory cytokines and the GPX4/SLC7A11/Nrf2 pathway. Full article
Show Figures

Graphical abstract

Back to TopTop