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 (3,269)

Search Parameters:
Keywords = apoptosis inducing factor-1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 3795 KB  
Article
Multi-Omics Analyses Reveal the Antifungal Mechanism of Phenazine-1-Carboxylic Acid Against Pseudogymnoascus destructans
by Zihao Huang, Shaopeng Sun, Zhouyu Jin, Yantong Ji, Jiaqi Lu, Ting Xu, Keping Sun, Zhongle Li and Jiang Feng
J. Fungi 2026, 12(1), 16; https://doi.org/10.3390/jof12010016 (registering DOI) - 25 Dec 2025
Abstract
White-nose syndrome (WNS) is an infectious disease of bats caused by the psychrophilic fungus Pseudogymnoascus destructans. Phenazine-1-carboxylic acid (PCA) is a microbial secondary metabolite with broad-spectrum antifungal activity. Previous studies show that PCA suppresses the growth of P. destructans at low concentrations, [...] Read more.
White-nose syndrome (WNS) is an infectious disease of bats caused by the psychrophilic fungus Pseudogymnoascus destructans. Phenazine-1-carboxylic acid (PCA) is a microbial secondary metabolite with broad-spectrum antifungal activity. Previous studies show that PCA suppresses the growth of P. destructans at low concentrations, yet its mechanism remains unclear. Here, we evaluated the in vitro antifungal activity of PCA. We then investigated its potential mechanism using physiological and biochemical assays, as well as integrated transcriptomic and metabolomic analyses. PCA showed effective antifungal activity against P. destructans (EC50 = 32.9 μg/mL). Physiological and biochemical assays indicated that PCA perturbed cell wall organization and increased membrane permeability, leading to leakage of intracellular contents. It also induced oxidative stress, DNA damage, and apoptosis. Multi-omics integration revealed that PCA markedly perturbed cell wall and membrane metabolism, virulence factor expression, and energy metabolism. It provoked oxidative stress while downregulating genes involved in the cell cycle, DNA replication, and repair. Together, these findings delineate the inhibitory effects of PCA on P. destructans in vitro, provide initial mechanistic insights into its antifungal action, and suggest that PCA merits further evaluation as a possible component of environmentally compatible strategies for WNS management. Full article
(This article belongs to the Section Fungal Genomics, Genetics and Molecular Biology)
Show Figures

Figure 1

21 pages, 3020 KB  
Article
Potential Anticancer Effect of Cannabis sativa L. Dichloromethane Extract Through Oxidative Stress-Related Pathways and the Inhibition of the Migration and Invasiveness of Human Breast Cancer Cells (MDA-MB-231 and MCF-7)
by Corinne Raïssa Ngnameko, Jacqueline Njikam Manjia and Motlalepula Gilbert Matsabisa
Int. J. Mol. Sci. 2026, 27(1), 152; https://doi.org/10.3390/ijms27010152 - 23 Dec 2025
Abstract
Breast cancer remains a leading cause of cancer-related morbidity and mortality globally, highlighting the urgent need for novel therapeutic strategies. This study investigates the molecular mechanisms underlying the anti-proliferative potential of Cannabis sativa dichloromethane extract (C. sativa DCM) on oxidative stress, apoptosis, [...] Read more.
Breast cancer remains a leading cause of cancer-related morbidity and mortality globally, highlighting the urgent need for novel therapeutic strategies. This study investigates the molecular mechanisms underlying the anti-proliferative potential of Cannabis sativa dichloromethane extract (C. sativa DCM) on oxidative stress, apoptosis, and invasion in human breast cancer cells. Key biomarkers, such as antioxidant enzymes (Superoxide Dismutase (SOD) and Glutathione (GSH)), the transcription factor Nrf2, apoptotic proteins (p53, caspase-8 and 9), metalloproteinase (MMP-1 and MMP-9), and Transforming Growth Factor Beta (TGF-β) were examined. Cytotoxicity was assessed using an MTT assay in the MDA-MB-231 and MCF-7 breast cancer cell lines, with comparisons to normal skin fibroblasts (HS27). Oxidative stress biomarkers were quantified using enzymatic assays and ELISA kits, while apoptotic and anti-metastatic factors were determined by Western blotting. Results demonstrated that C. sativa DCM extract induced significant cell death in a concentration-dependent manner, with IC50 values of 75.46 ± 0.132 μg/mL for MDA-MB-231 and 78.68 ± 0.50 μg/mL for MCF-7 cells. The extract decreased SOD and GSH levels while increasing p53 and caspase activity, confirming apoptosis activation. Additionally, C. sativa DCM inhibited migration and invasion by downregulating MMP-1, MMP-9, and TGF-β. The anti-proliferative potential of C. sativa DCM in breast cancer cells is mediated through a continuous biological pathway involving oxidative stress modulation, apoptotic signaling, and anti-invasive effects. Phytochemical analysis revealed terpenoids and steroids, including compounds like cannabidiol and tetrahydrocannabinol acid. These findings suggest that C. sativa DCM extract holds potential as an anti-breast cancer therapeutic and warrants further preclinical and clinical investigations. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

15 pages, 23101 KB  
Article
Identification of Differentially Expressed Genes and Molecular Pathways Involved in Primary Biliary Cholangitis Using RNA-Seq
by Min Yang, Xiaoyun Shen, Haitao Fu, Jie Lu and Fengying Li
Genes 2026, 17(1), 10; https://doi.org/10.3390/genes17010010 - 22 Dec 2025
Viewed by 117
Abstract
Objective: This study aims to investigate the functional role of lncRNA STX17-DT, which was previously found to be upregulated in peripheral blood mononuclear cells (PBMCs) of PBC patients, by examining its impact on gene expression and cellular behavior in a human monocyte [...] Read more.
Objective: This study aims to investigate the functional role of lncRNA STX17-DT, which was previously found to be upregulated in peripheral blood mononuclear cells (PBMCs) of PBC patients, by examining its impact on gene expression and cellular behavior in a human monocyte model. Methods: STX17-DT was overexpressed in THP-1 cells, which was assessed via plasmid transfection. Transcriptomic changes were analyzed by RNA sequencing, followed by comprehensive bioinformatics analyses including differential expression, functional enrichment, transcription factor network, and protein–protein interaction (PPI) analysis. Functional validation was performed using CCK-8 and TUNEL assays to assess proliferation and apoptosis, respectively. Results: Overexpression of STX17-DT led to 1973 differentially expressed genes (DEGs), with 1201 upregulated and 772 downregulated. Key upregulated genes included interferon-stimulated genes (e.g., interferon induced protein 44 like (IFI44L), interferon induced protein 44 (IFI44), guanylate binding protein 1(GBP1)) and chemokines (CCL4, CCL8). Upregulated DEGs were significantly enriched in immune-related pathways such as NF-κB signaling, Toll-like receptor signaling, TNF signaling, and cytokine–cytokine receptor interaction. Downregulated genes were involved in metabolic and signaling pathways such as PI3K–Akt, cAMP, and butanoate metabolism. Transcription factor analysis revealed significant alterations in regulators like Yes1 associated transcriptional regulator(YAP1), nuclear receptor subfamily 4 group A member 1(NR4A1), and MAF bZIP transcription factor B(MAFB). PPI network analysis suggested TNF, TLR4, TLR6, and STAT2 as central hubs. Functionally, STX17-DT overexpression enhanced THP-1 cell proliferation and significantly reduced apoptosis. Conclusions: STX17-DT promoted a pro-inflammatory transcriptomic profile and enhanced monocyte survival in our study, suggesting a potential role in PBC immunopathology. It may represent a potential biomarker and therapeutic target, particularly for patients with advanced disease or suboptimal response to ursodeoxycholic acid. Further studies in primary cells, animal models, and histological samples are warranted to validate its role in PBC pathogenesis. Full article
(This article belongs to the Section RNA)
Show Figures

Figure 1

16 pages, 26224 KB  
Article
Exploring the Protective Effect of Gastrodia elata Extract on D-Galactose-Induced Liver Injury in Mice Based on the PI3K/Akt Signaling Pathway
by Liu Han, Hongyu Zhai, Xiangyu Ma, He Li, Qiaosen Ren, Jiating Liu, Zhe Zhang, Xintong Li, Qiuyue Zhang and Xin Sun
Curr. Issues Mol. Biol. 2026, 48(1), 6; https://doi.org/10.3390/cimb48010006 - 20 Dec 2025
Viewed by 131
Abstract
In this research, we sought to methodically examine the protective effects of Gastrodia elata extract (GEE) on liver damage induced by D-galactose (D-gal) in mice and clarify the underlying mechanisms. The chemical composition of GEE was characterized using Ultra-Performance Liquid Chromatography–Tandem Mass Spectrometry [...] Read more.
In this research, we sought to methodically examine the protective effects of Gastrodia elata extract (GEE) on liver damage induced by D-galactose (D-gal) in mice and clarify the underlying mechanisms. The chemical composition of GEE was characterized using Ultra-Performance Liquid Chromatography–Tandem Mass Spectrometry (UPLC-MS/MS), while network pharmacology analysis was employed to predict potential molecular targets and signaling pathways. A mouse model of liver injury was established through daily intraperitoneal injection of D-gal over a 42-day period, during which the hepatoprotective efficacy of GEE was evaluated. Biochemical, histopathological, and molecular analyses were subsequently performed. UPLC-MS/MS identified ingredients such as amino acids, aromatic compounds, fatty acids, and terpenoids in GEE. A network pharmacology analysis enabled the identification of 272 common targets linked to GEE and liver damage, demonstrating notable enrichment within the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. In vivo experiments demonstrated that GEE effectively alleviated D-gal-induced body weight loss and elevated liver index values, alleviated hepatic histological damage, and reduced serum levels of Alanine Aminotransferase (ALT), Aspartate Aminotransferase (AST), and Alkaline Phosphatase (ALP). Furthermore, GEE enhanced the activities of the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT), decreased malondialdehyde (MDA) levels, and downregulated the mRNA expression of the pro-inflammatory cytokines Interleukin-6 (IL-6), Interleukin-1 beta (IL-1β), and Tumor Necrosis Factor-alpha (TNF-α). Western blot analysis confirmed that GEE activated the PI3K/Akt pathway, as evidenced by increased ratios of phosphorylated Phosphatidylinositol 3-kinase/Phosphatidylinositol 3-kinase (p-PI3K/PI3K) and phosphorylated AKT/Protein Kinase B (p-AKT/AKT); restored the B-cell lymphoma 2-associated X protein/B-cell lymphoma-2 (Bax/Bcl-2) balance; and reduced cyclin-dependent kinase inhibitor 1 (p21) expression. The results suggest that GEE protects against D-gal-induced liver damage by reducing oxidative stress, inhibiting inflammatory responses, and modulating apoptosis through the activation of the PI3K/Akt signaling pathway, providing support for its potential use in hepatoprotection. Full article
Show Figures

Figure 1

17 pages, 2171 KB  
Article
Targeting Redox Homeostasis and Cell Survival Signaling with a Flavonoid-Rich Extract of Bergamot Juice in In Vitro and In Vivo Colorectal Cancer Models
by Alessandro Maugeri, Paola De Cicco, Rebecca Amico, Martina Farina, Michele Navarra and Francesca Borrelli
Pharmaceutics 2026, 18(1), 7; https://doi.org/10.3390/pharmaceutics18010007 - 20 Dec 2025
Viewed by 137
Abstract
Background/Objectives: Colorectal cancer (CRC) is the second most common cause of cancer death worldwide. Evidence suggests that a polyphenol-rich diet may lower the risk of CRC. The aim of this study was to demonstrate the potential antitumor effects of a flavonoid-rich extract [...] Read more.
Background/Objectives: Colorectal cancer (CRC) is the second most common cause of cancer death worldwide. Evidence suggests that a polyphenol-rich diet may lower the risk of CRC. The aim of this study was to demonstrate the potential antitumor effects of a flavonoid-rich extract of bergamot juice (BJe) in both in vitro and in vivo CRC models, assessing the underlying mechanisms. Methods: CRC cells, among which HCT-116, have been employed to assess the fine mechanism of action of BJe, whereas a mouse model of azoxymethane (AOM)-induced CRC was exploited to appreciate the anti-cancer effects of BJe. Results: BJe inhibited the growth of several CRC cells, especially HCT-116. In this cell line, BJe induced apoptosis and blocked the cell cycle in the G1 phase, as well as modulated the gene expression of apoptosis- and cell cycle-related factors. Moreover, BJe prompted reactive oxygen species production and impaired mitochondrial membrane potential. In the nucleus of these cancerous cells, BJe induced DNA damage as confirmed by the raised levels of 8-oxo-2′-deoxyguanosine and phosphorylation of histone H2A.X. In mice with AOM-induced CRC, BJe was able to lower the number of aberrant crypt foci. Moreover, BJe reduced the percentage of mice bearing both polyps and tumors, as well as their number. Conclusions: Our study supports the role of BJe against CRC, providing knowledge on the underlying mechanism of action. Full article
Show Figures

Figure 1

24 pages, 1330 KB  
Review
Effects of Common Fig (Ficus carica L.) and Its Extracts on Certain Cancer Types: Focusing on the Mechanism of Action
by Elif Nisa Gökçen, Sevgi Gezici, Bence L. Raposa, Dávid Szép, Ferenc Budán and Duygu Ağagündüz
Int. J. Mol. Sci. 2026, 27(1), 56; https://doi.org/10.3390/ijms27010056 - 20 Dec 2025
Viewed by 129
Abstract
Cancer continues to be a global health problem due to high mortality rates and resistance to treatment. Since conventional chemotherapies cause serious side effects, interest in natural complementary therapies has increased. In this context, common fig (Ficus carica L.) (F. carica [...] Read more.
Cancer continues to be a global health problem due to high mortality rates and resistance to treatment. Since conventional chemotherapies cause serious side effects, interest in natural complementary therapies has increased. In this context, common fig (Ficus carica L.) (F. carica), which stands out with its rich phytochemical content, has been used in traditional medicine for a long time and attracts attention with its anticancer potential. The purpose of this review is to evaluate the biological effects of extracts obtained from different parts of the F. carica plant on cancer cells. Recent in vitro studies have shown that F. carica extracts suppress proliferation, induce apoptosis and reduce oxidative stress in various cancer cell lines. However, factors such as the plant part used, extraction method, dose and application time have caused differences in the results. In vivo studies are limited and there is no clinical study. Some studies report that high doses, especially latex, may cause toxic effects. F. carica extracts are promising against cancer. However, comprehensive in vivo and clinical studies with standardized extracts are needed to transfer this potential to clinical practice. Full article
(This article belongs to the Special Issue Molecular Insight into Plant Bioactive Compounds)
Show Figures

Figure 1

21 pages, 1710 KB  
Review
Modified mRNA-Based Therapeutic Strategies for Myocardial Ischemia–Reperfusion Injury
by Ting Cai and Xiang-Qun Yang
Int. J. Mol. Sci. 2026, 27(1), 55; https://doi.org/10.3390/ijms27010055 - 20 Dec 2025
Viewed by 220
Abstract
Ischemic heart disease (IHD), the leading causes of cardiovascular morbidity and mortality worldwide, is currently treated though revascularization strategies such as pharmacological thrombolysis, coronary artery bypass grafting (CABG), and percutaneous coronary intervention (PCI). However, the restoration of blood flow often induces cardiac dysfunction, [...] Read more.
Ischemic heart disease (IHD), the leading causes of cardiovascular morbidity and mortality worldwide, is currently treated though revascularization strategies such as pharmacological thrombolysis, coronary artery bypass grafting (CABG), and percutaneous coronary intervention (PCI). However, the restoration of blood flow often induces cardiac dysfunction, known as myocardial ischemia–reperfusion injury (MIRI). The pathogenesis of MIRI involves a complex, multifactorial process characterized by the interplay of diverse pathophysiological mechanisms, including oxidative stress, intracellular calcium overload, inflammatory cascade activation, apoptosis, autophagy, and microvascular endothelial dysfunction. In recent years, modified RNA (modRNA) technology has emerged as a novel therapeutic strategy for MIRI due to its enhanced molecular stability, reduced immunogenicity, and controllable transient protein expression. Studies have demonstrated that optimized modRNA delivery systems enable efficient, localized expression of therapeutic genes (e.g., antioxidant, anti-apoptotic, and pro-angiogenic factors) at injury sites, significantly mitigating MIRI-associated pathological damage. Nevertheless, significant challenges remain in clinical translation, such as delivery system targeting, transfection efficiency and cytotoxicity. This review focuses on recent advances in the development and application of modRNA-based delivery systems for MIRI treatment. Understanding the molecular mechanisms of MIRI and the structural characteristics and application of modRNA may encourage researchers to explore promising therapeutic modalities for addressing reperfusion-related cardiac injury. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

17 pages, 4240 KB  
Article
Topical Administration of Sitagliptin Prevents Retinal Neurodegeneration in a Model of Glaucoma Induced by Dexamethasone
by Patricia Bogdanov, Anna Duarri, David Sabater, María José Canz, Helena Isla-Magrané, Hugo Ramos, Anna Deàs-Just, Rafael Simó and Cristina Hernández
Int. J. Mol. Sci. 2026, 27(1), 48; https://doi.org/10.3390/ijms27010048 - 20 Dec 2025
Viewed by 115
Abstract
Glaucoma is a neurodegenerative disease characterized by progressive degeneration of optic nerve axons and loss of retinal ganglion cells (RGCs). Although elevated intraocular pressure (IOP) is a major risk factor, many patients develop glaucoma with normal IOP, highlighting the need for neuroprotective therapies. [...] Read more.
Glaucoma is a neurodegenerative disease characterized by progressive degeneration of optic nerve axons and loss of retinal ganglion cells (RGCs). Although elevated intraocular pressure (IOP) is a major risk factor, many patients develop glaucoma with normal IOP, highlighting the need for neuroprotective therapies. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, has shown beneficial effects in diabetes-induced retinal neurodegeneration. This study aimed to evaluate whether sitagliptin eye drops, previously effective in diabetes-induced retinal neurodegeneration, could prevent corticosteroid-induced glaucoma. Glaucoma was induced in mice by periocular injection of dexamethasone (DEX) once weekly for five weeks. Sitagliptin or vehicle eye drops were administered from day 14 to 35. Untreated mice served as controls. DEX treatment caused significant loss of RGC bodies and optic nerve axons compared to controls, which was prevented by sitagliptin eye drops (p < 0.001), without affecting IOP. Sitagliptin also inhibited DEX-induced activation of macroglia and microglia and prevented oligodendrocyte loss. Furthermore, it suppressed overexpression of galectin-3 and gamma-synuclein in the optic nerve head (ONH) (p < 0.001), key mediators of inflammation and apoptosis. Sitagliptin eye drops exert a potent neuroprotective effect against corticosteroid-induced glaucoma, supporting their potential as a novel therapeutic strategy for glaucoma. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Treatment of Retinal Diseases)
Show Figures

Graphical abstract

32 pages, 6067 KB  
Review
Molecular Regulation of FOXO1 and Its Pathophysiological Significance in Endometriosis: A Narrative Review
by Hiroshi Kobayashi, Hiroshi Shigetomi, Miki Nishio, Mai Umetani, Shogo Imanaka and Hiratsugu Hashimoto
Antioxidants 2026, 15(1), 3; https://doi.org/10.3390/antiox15010003 - 19 Dec 2025
Viewed by 217
Abstract
Background: Endometriosis is a chronic inflammatory disorder that affects approximately 10% of women of reproductive age and exhibits tumor-like characteristics such as invasion, recurrence, and hormone-dependent proliferation despite its benign nature. Its pathogenesis is thought to involve hormonal imbalance, oxidative stress, hypoxia, immune [...] Read more.
Background: Endometriosis is a chronic inflammatory disorder that affects approximately 10% of women of reproductive age and exhibits tumor-like characteristics such as invasion, recurrence, and hormone-dependent proliferation despite its benign nature. Its pathogenesis is thought to involve hormonal imbalance, oxidative stress, hypoxia, immune dysregulation, and epigenetic alterations. This review summarizes how these factors contribute to lesion formation through intracellular signaling pathways, with a particular focus on the role of the stress-responsive transcription factor Forkhead box O (FOXO1). Methods: A comprehensive literature search was conducted using PubMed and Google Scholar without temporal restriction. Results: FOXO1 is a transcription factor that integratively regulates decidualization, cellular senescence, autophagy, and apoptosis. In the normal endometrium, under mild stress or hormonal stimulation, FOXO1 induces decidualization-associated genes (PRL, IGFBP1) and antioxidant enzymes, thereby promoting differentiation and survival. In contrast, in endometriosis, activation of the PI3K/AKT signaling pathway and an estrogen-dominant environment suppress the nuclear activity of FOXO1, leading to apoptosis resistance, accumulation of senescent cells, and chronic inflammation through the senescence-associated secretory phenotype (SASP). Moreover, depending on the intensity and duration of oxidative, metabolic, and environmental stress, FOXO1 drives distinct cellular fates—including decidualization, senescence, and apoptosis—thus contributing to the persistence and progression of endometriotic lesions. Conclusion: Dysregulation of the FOXO1-dependent cellular fate–control network plays a central role in the development of endometriosis. Elucidating the molecular mechanisms governing FOXO1 activity and its nuclear dynamics will be crucial for a comprehensive understanding of disease progression and for the development of novel therapeutic strategies. Full article
Show Figures

Figure 1

23 pages, 8949 KB  
Article
Sunitinib Impairs Oral Mucosal Healing Through Endoplasmic Reticulum Stress-Mediated Keratinocyte Dysfunction
by Jiarui Wang, Lihang Shen, Shuo Chen, Xinyu Wang, Yang He and Yi Zhang
Cells 2026, 15(1), 1; https://doi.org/10.3390/cells15010001 - 19 Dec 2025
Viewed by 189
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a severe adverse event triggered by antiresorptive and/or anti-angiogenic agents, characterized by bone destruction, sequestrum formation, and refractory mucosal defects. Effective mucosal healing can be a critical factor for MRONJ prevention and treatment. While endoplasmic reticulum [...] Read more.
Medication-related osteonecrosis of the jaw (MRONJ) is a severe adverse event triggered by antiresorptive and/or anti-angiogenic agents, characterized by bone destruction, sequestrum formation, and refractory mucosal defects. Effective mucosal healing can be a critical factor for MRONJ prevention and treatment. While endoplasmic reticulum stress (ER stress) has been implicated in tissue repair, its role in MRONJ-associated mucosal healing impairment remains undefined. This study investigated the effects of the anti-angiogenic drug sunitinib on oral mucosal healing and its underlying mechanisms. A mouse model of palatal mucosal defects was established, RNA-seq, transmission electron microscopy, and morphological analyses were used to assess how sunitinib affects ER function during mucosal repair. Using human oral keratinocytes (HOKs), we further elucidated the subcellular mechanisms through which sunitinib influences cell proliferation, migration, cell cycle progression, tight junctions, and apoptosis via techniques such as qPCR, Western blotting, immunofluorescence, and flow cytometry. Our findings demonstrated that sunitinib might induce significant alterations in the morphology of the ER and mitochondria. Both in vivo and in vitro experiments revealed that sunitinib persistently activates the GRP78 (BIP)/PERK/ATF4/CHOP axis in HOKs. This sustained ER stress can inhibit keratinocytes migration and proliferation, disrupt tight junctions, and trigger the intrinsic mitochondrial apoptotic pathway, ultimately leading to impaired oral mucosal healing and barrier dysfunction. Critically, pharmacological inhibition of ER stress was shown to restore keratinocytes’ function and promote effective mucosal healing. These results indicated that targeting sunitinib-induced persistent ER stress might represent a promising therapeutic strategy to prevent and treat oral mucosal toxicity associated with this drug. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Figure 1

16 pages, 1526 KB  
Article
Role of Cytokines in Wound Healing Following Wound Catheter Analgesia in Rats
by Marija Lipar, Andrea Martinović, Tamara Nikuševa Martić, Tihana Kurtović, Jadranka Bubić Špoljar, Andrea Gelemanović, Marko Hohšteter, Lidija Medven Zagradišnik, Ivana Mihoković Buhin, Andrija Musulin, Višnja Nesek Adam, Božo Gorjanc, Slobodan Vukičević and Dražen Vnuk
Vet. Sci. 2025, 12(12), 1214; https://doi.org/10.3390/vetsci12121214 - 18 Dec 2025
Viewed by 107
Abstract
Background: Local analgesia administered through a wound catheter is widely used for postoperative pain control, yet its effects on wound healing remain incompletely understood. This study examined how levobupivacaine alone or combined with meloxicam or buprenorphine influences inflammatory markers, angiogenesis, apoptosis, and transforming [...] Read more.
Background: Local analgesia administered through a wound catheter is widely used for postoperative pain control, yet its effects on wound healing remain incompletely understood. This study examined how levobupivacaine alone or combined with meloxicam or buprenorphine influences inflammatory markers, angiogenesis, apoptosis, and transforming growth factor β1 (TGF-β1) expression during wound healing in rats. Methods: Thirty Sprague Dawley rats were assigned to five groups: control, saline, levobupivacaine (L), levobupivacaine/meloxicam (L/MEL), and levobupivacaine/buprenorphine (L/BUP). Treatments were administered via a wound catheter for three days. Blood and skin samples were collected before surgery and on days 3, 10, and 21. Results: Levobupivacaine combined with meloxicam or buprenorphine caused fluctuations in white blood cell counts, while albumin levels remained stable. Angiogenesis in the L/MEL group was markedly increased compared with the control, saline, and levobupivacaine-only groups, but the newly formed vessels exhibited consistently narrow lumina during the early healing phase. Caspase-3–positive cells were most numerous in L/MEL during inflammatory and proliferative phases, whereas delayed caspase-3 activation was observed in L/BUP. TGF-β1 expression peaked in both adjuvant groups on days 3 and 10. Conclusions: Meloxicam and buprenorphine increased TGF-β1 expression, but their vascular effects differed considerably. Meloxicam induced a marked increase in angiogenesis, but the newly formed vessels were structurally immature, displaying uniformly narrow lumina and poor architectural organisation, which led to their subsequent regression. In contrast, buprenorphine supported the formation of more mature vascular structures, characterised by wider vessel lumina and a more organised vascular network. These findings demonstrate that adjuvants used with levobupivacaine can significantly modify angiogenic and apoptotic responses and should be carefully considered when selecting multimodal local analgesia strategies after surgery. Full article
(This article belongs to the Section Veterinary Biomedical Sciences)
Show Figures

Figure 1

18 pages, 10030 KB  
Article
Ameliorative Effects of Vitamin E and Lutein on Hydrogen Peroxide-Triggered Oxidative Cytotoxicity via Combined Transcriptome and Metabolome Analysis
by Hongrui Lv, Yongji He and Shang Guo
Cells 2025, 14(24), 2020; https://doi.org/10.3390/cells14242020 - 18 Dec 2025
Viewed by 215
Abstract
Vitamin E and lutein both belong to food functional factors, which have cytoprotective potential and antioxidant effects. However, mechanism details at cell level remain scarce. In this study, HepG2 cells were utilized to inquire and compare the ameliorative effects of vitamin E and [...] Read more.
Vitamin E and lutein both belong to food functional factors, which have cytoprotective potential and antioxidant effects. However, mechanism details at cell level remain scarce. In this study, HepG2 cells were utilized to inquire and compare the ameliorative effects of vitamin E and lutein under H2O2-induced oxidative stress through a combined transcriptomic and metabolomic profiling, in addition to physiology and biochemistry determination. Cell cytotoxicity caused by H2O2 was ameliorated by vitamin E or lutein as evidenced by elevating cell viability and balancing the redox system. Vitamin E had greater efficacy on ameliorating oxidative cytotoxicity than lutein. Transcriptome data revealed that differentially expressed genes were mainly enriched in the transport-related, enzyme-related, and oxidative stress-related GO terms with vitamin E pretreatment. Extracellular organization-related, biological process-related, and apoptosis-related GO terms were meaningfully enriched with lutein pretreatment. Metabolome data showed that with vitamin E ameliorative effects, the disturbed metabolic pathways included thiamine metabolism, vitamin digestion and absorption, and ABC transporters. With lutein ameliorative effects, KEGG pathway analysis showed enrichment of amino sugar and nucleotide sugar metabolism, pyrimidine metabolism, and starch and sucrose metabolism. Collectively, our study provides essential insights into utilization of vitamin E and lutein as a potential supplement for effective therapy of disease associated with oxidative stress. Full article
Show Figures

Figure 1

26 pages, 5507 KB  
Article
A Fluid Dynamics-Model System for Advancing Tissue Engineering and Cancer Research Studies: Biological Assessment of the Innovative BioAxFlow Dynamic Culture Bioreactor
by Giulia Gramigna, Federica Liguori, Ludovica Filippini, Maurizio Mastantuono, Michele Pistillo, Margherita Scamarcio, Alessia Mengoni, Antonella Lisi, Giuseppe Falvo D’Urso Labate and Mario Ledda
Biomimetics 2025, 10(12), 848; https://doi.org/10.3390/biomimetics10120848 - 18 Dec 2025
Viewed by 184
Abstract
In this study, an innovative bioreactor, named BioAxFlow, particularly suitable for tissue engineering applications, is tested. Unlike traditional bioreactors, it does not rely on mechanical components to agitate the culture medium, but on the unique fluid-dynamics behaviour induced by the geometry of the [...] Read more.
In this study, an innovative bioreactor, named BioAxFlow, particularly suitable for tissue engineering applications, is tested. Unlike traditional bioreactors, it does not rely on mechanical components to agitate the culture medium, but on the unique fluid-dynamics behaviour induced by the geometry of the culture chamber, which ensures continuous movement of the medium, promoting the constant exposure of the cells to nutrients and growth factors. Using the human osteosarcoma cell line SAOS-2, the bioreactor’s ability to enhance cell adhesion and proliferation on polylactic acid (PLA) scaffolds, mimicking bone matrix architecture, is investigated. Cells cultured in the bioreactor showed significant improvement in cell growth and adhesion, compared to static cultures, and a more homogeneous cell distribution upon the scaffold surfaces, which is crucial for the development of functional tissue constructs. The bioreactor also preserves the osteogenic potential of SAOS-2 cells as assessed by the expression of key osteogenic markers. Additionally, it retains the tumorigenic characteristics of SAOS-2 cells, including the expression of pro-angiogenic factors and apoptosis-related genes. These results indicate that the BioAxFlow bioreactor could be an effective platform for tissue engineering and cancer research, offering a promising tool for both regenerative medicine applications and drug testing. Full article
Show Figures

Graphical abstract

18 pages, 15544 KB  
Article
FOXM1 Maintains Homeostasis and Self-Renewal in Wharton’s Jelly Mesenchymal Stem Cells
by Nan Li and Qiang Wu
Genes 2025, 16(12), 1517; https://doi.org/10.3390/genes16121517 - 18 Dec 2025
Viewed by 171
Abstract
Background: The transcription factor FOXM1 is a master regulator of the cell cycle and is implicated in various cell fate decisions. However, its functional role and regulatory network in human Wharton’s jelly mesenchymal stem cells (WJ-MSCs) remain poorly defined. This study aimed to [...] Read more.
Background: The transcription factor FOXM1 is a master regulator of the cell cycle and is implicated in various cell fate decisions. However, its functional role and regulatory network in human Wharton’s jelly mesenchymal stem cells (WJ-MSCs) remain poorly defined. This study aimed to elucidate the comprehensive function of FOXM1 in maintaining WJ-MSC stemness, proliferation, and survival, and to delineate the underlying molecular mechanisms. Methods: We used RNA Interference to knock down FOXM1 in WJ-MSCs. The phenotypic impacts were assessed through CCK-8, colony formation, migration, and flow cytometry assays. We analyzed transcriptomic changes using RNA-seq and verified the results through qRT-PCR and Western blotting. Results: Knockdown of FOXM1 significantly reduced the expression of core pluripotency factors (OCT4, SOX2, and NANOG), impairing stem cell identity and abolishing colony formation and migration capacities. Furthermore, FOXM1 deficiency induced G0/G1 phase cell cycle arrest, downregulated CCND1, and triggered apoptosis through a mechanism involving p53 accumulation, an increased BAX/BCL-2 ratio, and Caspase-3 activation. RNA-seq analysis further corroborated the systematic downregulation of cell cycle pathways and upregulation of apoptotic pathways upon FOXM1 deficiency. Conclusions: Our findings establish FOXM1 as a critical regulatory node that integrates stem cell identity with proliferative and survival signals to maintain WJ-MSC homeostasis. This study redefines FOXM1’s role in stem cell biology and provides a theoretical foundation for enhancing the therapeutic efficacy of WJ-MSCs by modulating this key factor. Full article
(This article belongs to the Special Issue Genetics and Epigenetics of Cellular Differentiation)
Show Figures

Figure 1

45 pages, 3742 KB  
Review
Metabolic Stress and Adaptation in Pancreatic β-Cells to Hypoxia: Mechanisms, Modulators, and Implications for Transplantation
by Jannat Akram, Prianna Menezes, Noorul Ibtesam Idris, Joanna Eliza Thomas, Radwan Darwish, Afrin Tania, Alexandra E. Butler and Abu Saleh Md Moin
Cells 2025, 14(24), 2014; https://doi.org/10.3390/cells14242014 - 17 Dec 2025
Viewed by 259
Abstract
Pancreatic β-cells are metabolically active endocrine cells with a high oxygen demand to sustain glucose-stimulated insulin secretion (GSIS). Hypoxia, arising from vascular disruption, islet isolation, or pathological states such as type 2 diabetes (T2D) and obstructive sleep apnoea (OSA), is a potent metabolic [...] Read more.
Pancreatic β-cells are metabolically active endocrine cells with a high oxygen demand to sustain glucose-stimulated insulin secretion (GSIS). Hypoxia, arising from vascular disruption, islet isolation, or pathological states such as type 2 diabetes (T2D) and obstructive sleep apnoea (OSA), is a potent metabolic stressor that impairs β-cell function, survival, and differentiation. At the molecular level, hypoxia-inducible factors (HIF-1α and HIF-2α) orchestrate transcriptional programs that shift β-cell metabolism from oxidative phosphorylation to glycolysis, modulate mitochondrial function, and regulate survival pathways such as autophagy and mitophagy. Crosstalk with nutrient-sensing mechanisms, redox regulation, growth factor signaling, and protein synthesis control further shapes adaptive or maladaptive outcomes. Hypoxia alters glucose, lipid, and amino acid metabolism, while mitochondrial dysfunction, oxidative stress, and inflammatory signaling contribute to progressive β-cell failure. Therapeutic strategies including incretin hormones, GABAergic signaling, erythropoietin, ChREBP inhibition, and activation of calcineurin–NFAT or oxygen-binding globins—offer potential to preserve β-cell viability under hypoxia. In islet transplantation, oxygen delivery technologies, ischemic preconditioning, mesenchymal stem cell–derived exosomes, and encapsulation systems show promise in mitigating hypoxic injury and improving graft survival. This review synthesizes current knowledge on β-cell responses to hypoxic stress, with emphasis on metabolic reprogramming, molecular signaling, and translational interventions, underscoring that targeted modulation of β-cell metabolism and oxygen handling can enhance resilience to hypoxia and improve outcomes in diabetes therapy and islet transplantation. Full article
(This article belongs to the Section Cellular Metabolism)
Show Figures

Figure 1

Back to TopTop