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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,234)

Search Parameters:
Keywords = apoptosis rates

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
35 pages, 1179 KB  
Review
Cannabidiol–Ion Channel Interactions Represent a Promising Preventive and Therapeutic Strategy in Hepatocellular Carcinoma
by María de Guadalupe Chávez-López, Arturo Avalos-Fuentes, Estrella del C. Cruz-Manzo, Pedro A. Aguirre-Arriaga, Benjamín Florán, Julio Isael Pérez-Carreón, Cecilia Bañuelos and Javier Camacho
Pathophysiology 2026, 33(1), 8; https://doi.org/10.3390/pathophysiology33010008 - 14 Jan 2026
Abstract
Hepatocellular carcinoma (HCC) is the main type of liver cancer and one of the malignancies with the highest mortality rates worldwide. HCC is associated with diverse etiological factors including alcohol use, viral infections, fatty liver disease, and liver cirrhosis (a major risk factor [...] Read more.
Hepatocellular carcinoma (HCC) is the main type of liver cancer and one of the malignancies with the highest mortality rates worldwide. HCC is associated with diverse etiological factors including alcohol use, viral infections, fatty liver disease, and liver cirrhosis (a major risk factor for HCC). Unfortunately, many patients are diagnosed at advanced stages of the disease and receive palliative treatment only. Therefore, early markers of HCC and novel therapeutic approaches are urgently needed. The endocannabinoid system is involved in various physiological processes such as motor coordination, emotional control, learning and memory, neuronal development, antinociception, and immunological processes. Interestingly, endocannabinoids modulate signaling pathways involved in cell survival, proliferation, apoptosis, autophagy, and immune response. Consistently, several cannabinoids have demonstrated potential antitumor properties in experimental models. The participation of metabotropic and ionotropic cannabinoid receptors in the biological effects of cannabinoids has been extensively described. In addition, cannabinoids interact with other targets, including several ion channels. Notably, several ion channels targeted by cannabinoids are involved in inflammation, proliferation, and apoptosis in liver diseases, including HCC. In this literature review, we describe and discuss both the endocannabinoid system and exogenous phytocannabinoids, such as cannabidiol and Δ9-tetrahydrocannabinol, along with their canonical receptors, as well as the cannabidiol-targeted ion channels and their role in liver cancer and its preceding liver diseases. The cannabidiol-ion channel association is an extraordinary opportunity in liver cancer prevention and therapy, with potential implications for several environments that are for the benefit of cancer patients, including sociocultural, public health, and economic systems. Full article
(This article belongs to the Section Cellular and Molecular Mechanisms)
Show Figures

Graphical abstract

21 pages, 7072 KB  
Article
Cold Shock Protein B as an Alternative to DMSO for Oocyte Vitrification
by Xinhai Wang, Jing Guo, Kaiyan Zhang, Yi Fang, Hongyu Liu, He Ding, Yang Lyu, Xin Ma and Wenfa Lyu
Antioxidants 2026, 15(1), 107; https://doi.org/10.3390/antiox15010107 - 14 Jan 2026
Abstract
Dimethyl sulfoxide (DMSO) is widely utilized in the vitrification of oocytes, but DMSO exhibits concentration-dependent toxicity, which can compromise oocyte developmental potential by disrupting key cellular processes. This study reports the first successful use of cold shock protein B (CspB protein) as a [...] Read more.
Dimethyl sulfoxide (DMSO) is widely utilized in the vitrification of oocytes, but DMSO exhibits concentration-dependent toxicity, which can compromise oocyte developmental potential by disrupting key cellular processes. This study reports the first successful use of cold shock protein B (CspB protein) as a substitute for DMSO in vitrification solutions for oocyte vitrification. Combining dynamics simulations and experimental validation, we demonstrated CspB’s ability to inhibit ice crystallization and recrystallization by stabilizing its position at the ice–water interface and reducing ice formation rates. Recombinant CspB was successfully expressed and shown to bind to the oolemma. In vitrification solutions, CspB (1–2 mg/mL) effectively reduced ice crystal size and enabled a significant reduction or complete replacement of DMSO. This strategy markedly improved the post-thaw survival rates of both mouse and bovine metaphase II (MII) oocytes. Furthermore, oocytes vitrified with an optimized formulation (15% ethylene glycol + 2 mg/mL CspB) exhibited developmental competence (cleavage and blastocyst rates), oxidative stress markers (ROS, GSH), mitochondrial function (membrane potential and content), and apoptosis levels (Caspase-3/9) comparable to those treated with a standard DMSO-containing system. Transcriptomic analysis revealed that CspB’s cryoprotection involves the modulation of the mTOR signaling pathway. This role was functionally confirmed, as activation of mTOR abolished CspB’s beneficial effects, reinstating oxidative damage, mitochondrial dysfunction, and apoptosis. Thus, the CspB protein replaces DMSO with direct ice crystal formation suppression and mTOR-mediated oxidative stress regulation. This study offers a protein-based alternative to conventional permeable cryoprotectants. This approach holds promise for improving reproductive biotechnologies across species. Full article
Show Figures

Figure 1

22 pages, 4486 KB  
Article
Astaxanthin as a Natural Photoprotective Agent: In Vitro and In Silico Approach to Explore a Multi-Targeted Compound
by Aida Lahmar, Balkis Abdelaziz, Nahla Gouader, Abir Salek, Imen Waer and Leila Chekir Ghedira
Sci. Pharm. 2026, 94(1), 8; https://doi.org/10.3390/scipharm94010008 - 13 Jan 2026
Abstract
Ultraviolet B radiation is a major cause of skin aging, cellular senescence, and inflammaging, mediated by the excessive production of reactive oxygen species (ROS) and induction of apoptosis. This study evaluated the photo-protective effects of astaxanthin, one of the strongest natural antioxidants, in [...] Read more.
Ultraviolet B radiation is a major cause of skin aging, cellular senescence, and inflammaging, mediated by the excessive production of reactive oxygen species (ROS) and induction of apoptosis. This study evaluated the photo-protective effects of astaxanthin, one of the strongest natural antioxidants, in UVB-treated keratinocytes. The antioxidant capacity of astaxanthin was evaluated using ABTS, DPPH, and NBT/riboflavin/SOD assays. HaCaT cells were exposed to 30 mJ/cm2 of UVB radiation. Photoprotective effects and accumulated ROS were evaluated in UVB-irradiated HaCaT cells by MTT and DCFH-DA assays. Nitric oxide levels were quantified using the Griess reagent. Apoptosis was assessed by dual staining using acridine orange/ethidium bromide, lysosomal integrity by acridine orange uptake, and cell migration by scratch assay. Cell adhesion was assessed on ECM-coated Nunc plates. Finally, we formulated a 0.5% astaxanthin-enriched cream. Astaxanthin mitigated UVB-induced damage by reducing intracellular ROS levels by 3.7-fold, decreasing nitric oxide production to 29.8 ± 7.7% at the highest concentration, and maintaining lysosomal integrity. The carotenoid significantly enhanced cell viability, increasing it from 60.64 ± 8.3% in UV-treated cells to 102.1 ± 3.22% at 40 µM. Moreover, treated cells showed a significant reduction (p < 0.001) in the apoptotic rate (37.7 ± 3.1 vs. 87.7 ± 3.8 in UVB-irradiated cells, as evidenced by reduced chromatin condensation and nuclear fragmentation. Astaxanthin also enhanced tissue repair, as evidenced by increased cell migration and adhesion to several extracellular matrix (ECM) proteins (poly-L-lysine, laminin, fibrinogen, vitronectin and collagen I). In silico molecular docking predicted strong binding affinities between astaxanthin and key cellular targets, including JAK2 (−9.9 kcal/mol, highest affinity), STAT3, FAK, COX-2, NF-k-B, MMP2, and MMP9. The formulated cream demonstrated an in vitro SPF of 7.2 ± 2.5. Astaxanthin acts as a multifunctional photoprotective compound, providing a strong rationale for its incorporation into cosmetic and dermatological formulations, as further supported by the successful formulation and in vitro SPF estimation of an astaxanthin-enriched cream. Full article
Show Figures

Figure 1

18 pages, 2338 KB  
Article
L1CAM Promotes Human Endometrial Cancer Via NF-κB Activation
by Hiroyuki Kurosu, Hiroshi Asano, Alaa-eldin Salah-eldin, Kazuya Hamada, Shugo Tanaka, Asuka Ishii, Issei Kawakita, Kentaro Kumagai, Kensuke Nakazono, Yuko Katayama, Rino Saito, Chihiro Terasaka, Sari Iwasaki, Satoshi Tanaka, Atsushi Niida, Hidemichi Watari and Koji Taniguchi
Cancers 2026, 18(2), 198; https://doi.org/10.3390/cancers18020198 - 8 Jan 2026
Viewed by 361
Abstract
Background/Objectives: Endometrial cancer is one of the most common gynecological malignancies, with increasing incidence and mortality rates, particularly in developed countries. L1 cell adhesion molecule (L1CAM) has been identified as a poor prognostic factor for human endometrial cancer; however, the molecular mechanisms [...] Read more.
Background/Objectives: Endometrial cancer is one of the most common gynecological malignancies, with increasing incidence and mortality rates, particularly in developed countries. L1 cell adhesion molecule (L1CAM) has been identified as a poor prognostic factor for human endometrial cancer; however, the molecular mechanisms underlying its role in tumor progression remain unclear. Methods: We investigated the biological significance of L1CAM in human endometrial cancer using multiple cell lines. Functional analyses, including cell proliferation, cell cycle, and apoptosis assays, were performed after L1CAM knockdown or overexpression. Results: L1CAM promoted the transition of endometrial cancer cells from the G0/G1 phase and enhanced cell proliferation. L1CAM knockdown inhibited NF-κB signaling by reducing NF-κB (p65) phosphorylation and downregulating the expression of downstream targets such as TNF. Overexpression of constitutively active IKKβ restored the proliferation defect caused by L1CAM knockdown, supporting the role of NF-κB as a key downstream effector of L1CAM. Immunohistochemical analysis revealed a significant correlation between L1CAM expression and nuclear NF-κB (p65) positivity rates in human patient samples. Furthermore, combination therapy with cisplatin and an IKK inhibitor enhanced the anti-proliferative effect. Conclusions: Our study demonstrated that L1CAM promotes proliferation and chemotherapy resistance in human endometrial cancer through activation of the NF-κB signaling pathway. Therapeutic strategies targeting the L1CAM-NF-κB pathway may represent a promising treatment option for improving prognosis in L1CAM-positive human endometrial cancer. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

21 pages, 3994 KB  
Article
Elucidating the Mechanism of the Liqi Yangyin Formula in Treating Depression–Constipation Comorbidity: An Integrative Approach Using Network Pharmacology and Experimental Validation
by Lianjie Xu, Shun Seng Ong, Xiaoyue Deng, Yunzhi Qian, Zhao Tang, Ming Li and Tianshu Xu
Pharmaceuticals 2026, 19(1), 106; https://doi.org/10.3390/ph19010106 - 7 Jan 2026
Viewed by 231
Abstract
Background: The traditional formula Liqi Yangyin (LQYY) has shown clinical and preclinical efficacy for depression with constipation, yet its molecular mechanisms remain incompletely defined. This study aimed to elucidate its mechanisms using an integrative approach. Methods: Constituents of LQYY were profiled [...] Read more.
Background: The traditional formula Liqi Yangyin (LQYY) has shown clinical and preclinical efficacy for depression with constipation, yet its molecular mechanisms remain incompletely defined. This study aimed to elucidate its mechanisms using an integrative approach. Methods: Constituents of LQYY were profiled by UPLC-MS/MS and integrated with network pharmacology and molecular docking to identify brain-accessible components and putative targets. A chronic unpredictable mild stress (CUMS) model was used for experimental validation. Outcomes included behavioral tests (sucrose preference test, open field test, and forced swimming test), gastrointestinal indices, including fecal water content, time of first black stool, and intestinal propulsion rate, histopathology of the prefrontal cortex (PFC) and colon, TUNEL staining, NeuN immunofluorescence, Western blotting, and qRT-PCR. Results: LQYY attenuated CUMS-induced weight loss and depressive-like behaviors and improved intestinal transit metrics. It reduced neuronal apoptosis in the PFC and ameliorated colonic injury. Mechanistically, docking and enrichment analyses highlighted hub targets (STAT3, AKT1, ESR1, IL-6, TNF, TP53) and the JAK/STAT pathway. In vivo, LQYY decreased IL-6, TNF-α, ESR1, TP53, and STAT3, and increased AKT1 in the PFC and colon; it also reduced the TUNEL-positive rate and restored NeuN labeling, upregulated Bcl-2, and downregulated p-JAK2/JAK2 and p-STAT3/STAT3 ratios, and the expression of Bax and cleaved-caspase-3 in the PFC, consistent with the suppression of pro-inflammatory and apoptotic signaling. Conclusions: LQYY exerts antidepressant and pro-motility effects in CUMS mice by modulating JAK2/STAT3-centered networks and inhibiting neuronal apoptosis, thus supporting a multi-component, multi-target strategy for treating depression with constipation, and providing a defined molecular hypothesis for future investigation. Full article
Show Figures

Graphical abstract

46 pages, 2310 KB  
Review
The Role of Natural Compounds in Bladder Urothelial Carcinoma Treatment
by Hangfei Jiang, Yueyin Chen, Xi Zeng, Rui Yang, Feng Zhang, Huanling Zhang, Erxiang Zhang, Xuzhang Wu, Deye Yan and Chunping Yu
Int. J. Mol. Sci. 2026, 27(2), 596; https://doi.org/10.3390/ijms27020596 - 7 Jan 2026
Viewed by 118
Abstract
Bladder urothelial carcinoma (BUC) ranks among the most common malignant tumors of the urinary system, with alarmingly high incidence and mortality rates. Current clinical treatments face challenges such as strong chemotherapy resistance and limited response rates to immunotherapy, creating an urgent need for [...] Read more.
Bladder urothelial carcinoma (BUC) ranks among the most common malignant tumors of the urinary system, with alarmingly high incidence and mortality rates. Current clinical treatments face challenges such as strong chemotherapy resistance and limited response rates to immunotherapy, creating an urgent need for novel alternative therapies. Natural products, characterized by multi-targeted antitumor activity, low toxicity, and broad availability, have emerged as highly promising adjunctive or alternative strategies in cancer treatment. Extensive research has elucidated the antitumor activities of natural products, including inhibition of cancer cell proliferation, induction of apoptosis, and modulation of the immune microenvironment. What’s more, their bioactive components, such as terpenoids and polyphenols, can synergistically enhance therapeutic efficacy while reducing toxicity risks associated with traditional therapies. This review will examine the roles of terpenoids, phenolics, alkaloids, and other natural products in BUC treatment, to provide directions for future research. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

19 pages, 1678 KB  
Review
Role of Extracellular Vesicles in Abdominal Aortic Aneurysm: Pathophysiology, Biomarkers, and Therapeutic Potentials
by Kazuki Takahashi, Yusuke Yoshioka, Naoya Kuriyama, Shinsuke Kikuchi, Nobuyoshi Azuma and Takahiro Ochiya
Int. J. Mol. Sci. 2026, 27(2), 567; https://doi.org/10.3390/ijms27020567 - 6 Jan 2026
Viewed by 125
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease. Although AAA is generally asymptomatic, the mortality rate remains very high once rupture occurs, even with successful treatment. The pathophysiology of AAA involves inflammatory cell infiltration, smooth muscle cell apoptosis, and extracellular matrix degradation. However, [...] Read more.
Abdominal aortic aneurysm (AAA) is a life-threatening disease. Although AAA is generally asymptomatic, the mortality rate remains very high once rupture occurs, even with successful treatment. The pathophysiology of AAA involves inflammatory cell infiltration, smooth muscle cell apoptosis, and extracellular matrix degradation. However, there are various unclear aspects of pathophysiology due to cellular heterogeneity and multifactorial disease. Moreover, there are no blood biomarkers or available pharmacological drugs for AAA. Extracellular vesicles (EVs) are lipid bilayer particles released from every type of cell for intercellular communication. EVs include proteins, DNA, RNA (mRNA, microRNA), and lipids. EV cargos are delivered to recipient cells and modulate their biological effects. Although fewer studies have investigated EVs in AAA than in other cardiovascular diseases with similar molecular mechanisms, recent research indicates that EVs play a significant role in AAA development. Further research on EVs and AAA will contribute to the elucidation of AAA pathophysiology and the development of novel pharmacological drugs. In this review, we summarize the EV-associated pathophysiology, EV-based biomarkers, and EV-based treatment strategies in AAA. We also discuss the prospects for EVs research in AAA. Full article
(This article belongs to the Special Issue Exosomes—3rd Edition)
Show Figures

Figure 1

17 pages, 2315 KB  
Article
Studies of Akt/mTOR–Autophagy–Apoptosis Crosstalk in Human Gingival Fibroblasts Around New Digital Processed Dental Composites
by Florentina Rus, Radu Radulescu, Alexandra Popa, Monica Musteanu, Melis Izet, Corina Muscurel, Lucian Toma Ciocan, Sebastian-Andrei Bancu, Marina Imre and Alexandra Ripszky
Appl. Sci. 2026, 16(1), 532; https://doi.org/10.3390/app16010532 - 5 Jan 2026
Viewed by 127
Abstract
Introduction: Incomplete polymerization of in vivo composite resins (CR) poses a significant problem, with monomer-to-polymer conversion rates ranging from around 60 to 75%. Furthermore, oxygen exposure hampers polymerization in the surface layers. This research aims to evaluate the autophagy-inducing potential of three types [...] Read more.
Introduction: Incomplete polymerization of in vivo composite resins (CR) poses a significant problem, with monomer-to-polymer conversion rates ranging from around 60 to 75%. Furthermore, oxygen exposure hampers polymerization in the surface layers. This research aims to evaluate the autophagy-inducing potential of three types of CRS and to explore the role of the Akt/mTOR–autophagy–apoptosis crosstalk in composite resin-induced autophagy. The study uses human gingival fibroblasts and three composite materials (M1 and M2, which are 3D printed, and M3, which is milled). Materials and Methods: SEM analysis was performed on the dental materials, and cells kept in contact for 24 h were subjected to tests including the following: MTT, LDH, NO, immunological detection of proteins involved in autophagy and apoptosis, as well as immunofluorescence tests (Annexin V and nucleus; mitochondria and caspase 3/7; detection of autophagosomes). Results: The results showed statistically significant decreases in cell viability with M1 and M2, linked to increases in cytotoxicity and oxidative stress (LDH and NO). Using multiplex techniques, significant increases in glycogen synthase kinase 3 beta (GSK3b) protein were observed in both M1 and M2; a decrease in mTOR (mechanistic target of rapamycin) expression was noted in M1 and M3. Immunofluorescence tests revealed an increase in Annexin V across all materials studied, and an increase in autophagosomes in M1 and M2, whereas a decrease was observed in M3. Conclusions: The relationship between apoptosis and autophagy is highly complex, indicating they may occur sequentially, coexist, or be mutually exclusive. Understanding this complex interplay can help in designing new 3D-printing protocols and monomer compositions to prevent autophagy imbalance. Full article
(This article belongs to the Special Issue Advanced Biomaterials: Characterization and Applications)
Show Figures

Figure 1

18 pages, 5314 KB  
Article
NGR1 Pretreatment Enhances the Therapeutic Efficacy of Transplanting Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells for Myocardial Infarction
by Hao Cai, Meng-Ying Huang, Fang-Fang Mou, Qiang-Li Wang, Zhi-Rong Luo, Ping-Ping Lu, Bao-Nian Liu, Liang Hu and Hai-Dong Guo
Int. J. Mol. Sci. 2026, 27(1), 475; https://doi.org/10.3390/ijms27010475 - 2 Jan 2026
Viewed by 214
Abstract
Human induced pluripotent stem cells (hiPSCs) offer significant potential for differentiation and research applications in cardiovascular diseases. When induced differentiated hiPSC-derived cardiomyocytes (hiPSC-CMs) are transplanted into the infarcted myocardial region, they exhibit extremely low survival rates and unsatisfactory therapeutic effects due to ischemia, [...] Read more.
Human induced pluripotent stem cells (hiPSCs) offer significant potential for differentiation and research applications in cardiovascular diseases. When induced differentiated hiPSC-derived cardiomyocytes (hiPSC-CMs) are transplanted into the infarcted myocardial region, they exhibit extremely low survival rates and unsatisfactory therapeutic effects due to ischemia, hypoxia, and immune inflammation in the surrounding environment. To address this issue, we used Panax notoginseng saponin R1 (NGR1), which has demonstrated significant protective effects in prior research, to pretreat hiPSC-CMs before transplantation. Utilizing an in vitro H2O2 oxidative stress model and a nude mouse myocardial infarction (MI) model, we investigated the mechanism through which NGR1 pretreatment enhances the therapeutic efficacy of hiPSC-CM transplantation. The results revealed that the hiPSC-CMs expressed cTnT. NGR1 did not promote the proliferation of hiPSC-CMs but instead induced elevated levels of p-Akt protein in these cells. Compared to hiPSC-CM transplantation alone, transplantation of hiPSC-CMs pretreated with NGR1 exhibited higher ejection fraction (EF) and fractional shortening (FS) values, along with reduced infarct size and collagen deposition. Additionally, there were more HNA-positive cardiomyocytes in the cardiac tissue, fewer TUNEL-positive signals, and increased VWF-positive and Lyve1-positive signals. Furthermore, the gene expression levels of VEGFC, IGF-1, and SDF-1 were higher. Therefore, NGR1 pretreatment improves the survival of transplanted hiPSC-CMs in tissues, reduces myocardial apoptosis, enhances cardiac function, decreases infarct size and collagen deposition, promotes angiogenesis and lymphangiogenesis, and stimulates paracrine secretion. Full article
(This article belongs to the Special Issue Enhancing Stem Cell Grafting in Tissue Regeneration and Repair)
Show Figures

Figure 1

20 pages, 13115 KB  
Article
Apigenin Alleviates Zearalenone-Induced Oxidative Stress and Apoptosis in Swine Testis Cells Through the Wnt Signaling Pathway
by Chenyun Guo, Yidan Zhang, Yiwei Wang, Yile Sun, Haoze Ning, Jiaxin Gao, Fei Guo, Pengyun Ji, Lu Zhang, Guoshi Liu and Bingyuan Wang
Antioxidants 2026, 15(1), 42; https://doi.org/10.3390/antiox15010042 - 29 Dec 2025
Viewed by 284
Abstract
The mycotoxin zearalenone (ZEN), commonly found in contaminated food and feed, poses a significant threat to human and animal health, particularly to reproductive function. Mitigating its toxicity represents a critical research priority in food safety. Apigenin (AP) is a naturally occurring dietary flavonoid [...] Read more.
The mycotoxin zearalenone (ZEN), commonly found in contaminated food and feed, poses a significant threat to human and animal health, particularly to reproductive function. Mitigating its toxicity represents a critical research priority in food safety. Apigenin (AP) is a naturally occurring dietary flavonoid with phytoestrogenic properties and exhibits diverse pharmacological activities. In this study, we investigated the protective effects of AP against ZEN-induced apoptosis and oxidative stress in Swine Testis (ST) cells and elucidated its underlying mechanisms. The identity of ST cells was verified via RT-PCR and agarose gel electrophoresis. ST cells were treated with 40 μM ZEN and 1 μM and 0.1 μM AP for 24 h. Cell viability was detected via CCK8 and EdU assays, cytotoxicity was evaluated via LDH assay, cell cycle and apoptosis levels were analyzed via flow cytometry, and the mechanism by which AP alleviated the damage caused by ZEN to ST cells was preliminarily revealed using RNA-Seq technology. Further, the expression levels of related genes and proteins were detected by qRT-PCR and Western blot. Our results show that 1 μM or 0.1 μM AP effectively attenuated the cytotoxicity induced by 40 μM ZEN in ST cells, as evidenced by restored cell viability, reduced the LDH level, normalized cell cycle progression, reduced apoptotic rates, and enhanced antioxidant capacity. RNA-Seq analysis was coupled with molecular validation and used to elucidate the mechanisms underlying AP-mediated protection against ZEN-induced cellular injury. It is shown that ZEN suppressed the expression of LRP5, a pivotal gene in the Wnt signaling pathway, along with its downstream effector c-Myc. Conversely, treatment with 1 μM or 0.1 μM AP upregulated the expression of LRP5, iASPP, and TRAF2 at both transcriptional and translational levels. Importantly, the protective effects of AP were abrogated with IWR-1-endo, a specific Wnt pathway inhibitor, confirming pathway dependency. Collectively, our findings show that AP alleviates ZEN-induced oxidative stress and apoptosis in ST cells through the upregulation of LRP5 and subsequent activation of the Wnt signaling pathway. This study provides molecular evidence supporting the potential clinical application of AP as a preventive agent against ZEN-induced reproductive toxicity. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

17 pages, 2108 KB  
Article
Effects of Insulin-like Growth Factor I and Follicular Fluid on In Vitro Growth of Cultured Oocytes
by Yunfei Diao, Dengrong Zhai, Yunsu Wu, Puyuan Ai, Shuxuan Liu and Xiaoxia Li
Biology 2026, 15(1), 46; https://doi.org/10.3390/biology15010046 - 26 Dec 2025
Viewed by 198
Abstract
Oocyte-granulosa cell complexes (OGCs) cultivation is crucial for advancing reproductive biotechnology but remains incomplete and needs further optimization. Insulin-like growth factor-I (IGF-I) regulates granulosa cell proliferation and apoptosis, and numerous studies have confirmed its role in promoting ovarian follicle development. Porcine follicular fluid [...] Read more.
Oocyte-granulosa cell complexes (OGCs) cultivation is crucial for advancing reproductive biotechnology but remains incomplete and needs further optimization. Insulin-like growth factor-I (IGF-I) regulates granulosa cell proliferation and apoptosis, and numerous studies have confirmed its role in promoting ovarian follicle development. Porcine follicular fluid (PFF) contains factors beneficial for oocyte growth, which may enhance oocyte development. To investigate whether IGF-I and PFF improve the in vitro culture efficiency of porcine OGCs, we cultured OGCs with IGF-I (0, 10, 50, 100 ng/mL) and PFF (from 3 to 6 mm follicles) at concentrations of 0, 2.5%, 5%, 10%, respectively. The results revealed that 50 and 100 ng/mL IGF-I significantly increased the antrum formation rate of OGCs (from 61.11 ± 7.35% to 88.89 ± 7.35%) and diameter growth of oocytes (from 108.77 ± 0.27 µm to 114.94 ± 0.58 and 113.29 ± 0.50 µm, respectively). However, only the 50 ng/mL group, but not the 100 ng/mL group, significantly improved the maturation rate (38.13 ± 3.77% vs. 25.00 ± 3.27%, p < 0.05) of oocytes. Additionally, 50 ng/mL IGF-I downregulated BAX (a pro-apoptotic gene) and upregulated BCL-2 (an anti-apoptotic factor) in granulosa cells, ultimately reducing apoptosis. In contrast, none of the PFF doses used in this study induced the formation of enclosed antrum-like structures in OGCs, nor did they significantly enhance their in vitro development. Our findings demonstrate that 50 ng/mL IGF-I effectively promotes the in vitro growth of porcine early antral follicle-derived OGCs by reducing apoptosis, whereas tested PFF concentrations had no beneficial effects and induced abnormal granulosa cell growth. How PFF modulates the adherent and spreading growth of granulosa cells has not been fully elucidated and requires further clarification. Full article
(This article belongs to the Section Developmental and Reproductive Biology)
Show Figures

Figure 1

23 pages, 6790 KB  
Article
Sitagliptin Potentiates the Anticancer Activity of Doxorubicin Through ROS-Driven Apoptosis and MMP/TIMP Regulation in HeLa Cells
by Aşkın Evren Güler, Mehmet Cudi Tuncer and İlhan Özdemir
Pharmaceutics 2026, 18(1), 38; https://doi.org/10.3390/pharmaceutics18010038 - 26 Dec 2025
Viewed by 312
Abstract
Background/Objectives: Cervical cancer remains a major global health challenge, and treatment resistance limits the long-term success of chemotherapy. Drug repurposing strategies offer new opportunities for improving therapeutic outcomes by combining existing agents with established chemotherapeutics. Sitagliptin, a DPP-4 inhibitor commonly used in [...] Read more.
Background/Objectives: Cervical cancer remains a major global health challenge, and treatment resistance limits the long-term success of chemotherapy. Drug repurposing strategies offer new opportunities for improving therapeutic outcomes by combining existing agents with established chemotherapeutics. Sitagliptin, a DPP-4 inhibitor commonly used in type 2 diabetes, has recently gained attention for its potential anticancer effects. This study aimed to investigate the cytotoxic, apoptotic, and anti-metastatic effects of sitagliptin and doxorubicin, individually and in combination, on human cervical cancer cells (HeLa), and to determine whether their combined use exerts a synergistic anticancer effect. Methods: HeLa cells were treated for 48 h with increasing concentrations of sitagliptin, doxorubicin, or their combination. Cell viability was assessed using the MTT assay. Apoptosis was evaluated by Annexin V-FITC/PI staining and caspase-8/9 activity assays. Synergy was quantified using the Chou–Talalay method, and Combination Index (CI) values were used to determine synergistic interactions. Intracellular ROS levels were measured using the DCFDA assay. Migration and invasion capacities were analyzed using wound healing and Transwell assays. MMP-1, MMP-2, TIMP-1, and TIMP-2 levels were quantified via ELISA with normalization to viable cell counts. Gene expression levels of PI3K/Akt and MAPK/ERK pathway components were measured by qRT-PCR. Bioinformatic analyses (STRING, GeneMANIA, GO, KEGG) were performed to identify common molecular targets and enriched pathways affected by both agents. Results: The combination of sitagliptin and doxorubicin significantly reduced cell viability and demonstrated a synergistic interaction (CI < 1). Combined treatment induced a marked increase in ROS production and significantly elevated apoptosis rates compared to monotherapies. Caspase-8 and caspase-9 activities were also higher in the combination group. Migration and invasion assays revealed substantial suppression of cell motility and invasive capacity. After normalization to viable cell numbers, MMP and TIMP reductions remained significant, confirming true biological inhibition rather than cell-death–related artifacts. qRT-PCR analyses showed downregulation of Akt and ERK expression, indicating suppression of key survival and proliferation pathways. Bioinformatic analyses supported these findings by highlighting enrichment in apoptotic, oxidative stress, and metastasis-related pathways. Conclusions: Sitagliptin enhances the anticancer efficacy of doxorubicin by amplifying ROS-mediated apoptosis, inhibiting migration and invasion, and modulating PI3K/Akt and MAPK/ERK signaling in cervical cancer cells. The combination exhibits a clear synergistic effect and demonstrates strong potential as a supportive therapeutic strategy. These findings warrant further in vivo and clinical-level investigations to evaluate the translational applicability of sitagliptin in cervical cancer therapy. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

16 pages, 5893 KB  
Article
Co-Exposure of Microplastics and Avermectin at Environmental-Related Concentrations Caused Severe Heart Damage Through ROS-Mediated MAPK Signaling in Larval and Adult Zebrafish
by Guanghua Xiong, Min Lu, Yaxuan Jiang, Huangqi Shi, Jinghong Liu, Xinjun Liao, Huiqiang Lu, Yong Liu and Gaoxiao Xu
Toxics 2026, 14(1), 24; https://doi.org/10.3390/toxics14010024 - 25 Dec 2025
Viewed by 325
Abstract
The widespread presence of polystyrene microplastics (PS-MPs) and agricultural pollutants such as avermectin (AVM) in aquatic environments poses a significant threat to aquatic organisms. However, the combined toxic effect of PS-MPs and AVM on cardiac development remains poorly understood. This study aimed to [...] Read more.
The widespread presence of polystyrene microplastics (PS-MPs) and agricultural pollutants such as avermectin (AVM) in aquatic environments poses a significant threat to aquatic organisms. However, the combined toxic effect of PS-MPs and AVM on cardiac development remains poorly understood. This study aimed to investigate the cardiac toxicity of AVM co-exposed with two sizes of MPs (large MPs, LMPs, 20 µm; small MPs, SMPs, 80 nm) in both larval and adult zebrafish. Firstly, under the co-exposure conditions of MPs and AVM, we observed significant cardiac developmental toxicity, including decreased survival rate, body length, and hatching rate, as well as a significant reduction in the number of myocardial cells. Secondly, the number of neutrophils and antioxidant enzyme activities such as CAT and SOD were greatly decreased, while inflammatory cytokines such as TNF-α and IL8 were significantly increased after co-exposure in larval zebrafish. Thirdly, there was severe disorganization of cardiomyocytes and interstitial edema in adult zebrafish hearts under the co-exposure by histopathological examination. Our results suggest that cardiomyocyte proliferation was suppressed, but heart apoptosis level and anti-apoptotic genes were significantly increased in the AVM+MPs co-exposure. Additionally, transcriptome sequencing and bioinformatics analysis revealed that significant changes in differentially expressed genes in the AVM+SMPs co-exposure group, particularly in the processes related to oxidation–reduction, inflammatory response, and the MAPK signaling pathway in the adult zebrafish heart. Furthermore, our pharmacological experiments demonstrated that inhibiting ROS and blocking the MAPK signaling pathway could partially rescue the heart injury induced by AVM and MPs co-exposure in both larval and adult zebrafish. In summary, this study suggested that co-exposure to AVM and MPs could induce heart toxicity mainly via the ROS-mediated MAPK signaling pathway in zebrafish. The information provided important insights into the potential environmental risk of microplastic and pesticide co-exposure on aquatic ecosystems. Full article
Show Figures

Graphical abstract

27 pages, 12658 KB  
Article
Promoting Drp1-Mediated Mitochondrial Division in Nickel Nanoparticles-Induced Reproductive Toxicity in GC-2 Cells
by Liya Qiao, Zhimin Tong, Yabing Xu, Chunliu Guan, Geyu Liang and Lu Kong
Nanomaterials 2026, 16(1), 34; https://doi.org/10.3390/nano16010034 - 25 Dec 2025
Viewed by 272
Abstract
Male reproductive disorders and declining fertility rates play an important role in birth rates, and their impact on future populations makes them one of the most serious public health issues of this century. Defects in spermatogenesis are the most common manifestation of male [...] Read more.
Male reproductive disorders and declining fertility rates play an important role in birth rates, and their impact on future populations makes them one of the most serious public health issues of this century. Defects in spermatogenesis are the most common manifestation of male infertility, and exposure to environmental pollutants has been suggested as a potential cause. Nanomaterials, due to their unique physicochemical properties and widespread application, have raised growing concerns about their potential reproductive toxicity. Studies have shown that nickel nanoparticles (Ni NPs) have reproductive toxicity in male rats and mice, especially sperm damage. This study aimed to explore the male reproductive toxicity of Ni NPs and the role of mitochondrial fission in mouse spermatocytes (GC-2). Our results showed that Ni NPs induced the damage of mitochondrial structure and function in GC-2 cells and disrupted intramitochondrial homeostasis, thereby resulting in enhanced Dynamin-related protein 1(Drp1)-mediated mitochondrial fission and cell apoptosis, along with aggravated cytotoxicity and obvious reproductive toxicity. The mitochondrial division inhibitor 1(Mdivi-1) and lentiviral-transfected low expression of Dnm1l can significantly alleviate the germ cell toxicity caused by Ni NPs, suggesting a certain therapeutic effect. The novelty of this study lies in its systematic demonstration that Drp1-mediated mitochondrial division is a core pathogenic mechanism of Ni NP-induced male reproductive toxicity, and the validation of both pharmacological inhibition and genetic silencing as effective intervention strategies. Therefore, this study offers a reference for expanding the reproductive toxicity effect of Ni NPs and potential molecular mechanisms and provides an important basis for finding potential targets and treatment of Ni NPs. Full article
(This article belongs to the Special Issue Progress of Emerging Nanomaterials in Ecotoxicity and Biotoxicity)
Show Figures

Graphical abstract

18 pages, 4545 KB  
Article
Citrate Synthase Knockdown Suppresses Cell Proliferation and Induces Apoptosis in Select Human Cancer Cell Lines
by Xiaoxiao Zhang, Siyu Qu, Huanhuan Zhong, Yulu Yang, Bo Cheng and Yan Zeng
Int. J. Mol. Sci. 2026, 27(1), 83; https://doi.org/10.3390/ijms27010083 - 21 Dec 2025
Viewed by 294
Abstract
Citrate synthase (CS) catalyzes the first reaction in the tricarboxylic acid (TCA) cycle and is one of the rate-limiting and regulatory enzymes of the TCA cycle. How CS influences human cells beyond its direct roles in carbohydrate metabolism and energy production is poorly [...] Read more.
Citrate synthase (CS) catalyzes the first reaction in the tricarboxylic acid (TCA) cycle and is one of the rate-limiting and regulatory enzymes of the TCA cycle. How CS influences human cells beyond its direct roles in carbohydrate metabolism and energy production is poorly understood. In this study, we used RNA interference (RNAi) to knockdown CS expression in three diverse human cancer cell lines, HCT116, HT-1080, and HepG2, and assessed changes in their cellular behaviors. In all three cell lines, the loss of CS led to are duction in cell proliferation, increased apoptosis, lower mitochondrial membrane potentials, higher reactive oxygen species (ROS) production, and reduced ATP levels. We then performed transcriptome analyses in the three cell lines, identified pathways related to the cell cycle and apoptosis that might elucidate the mechanisms underlying those cellular changes, and further verified the mRNA expression changes in specific genes associated with the apoptotic pathways. Taken together, our results suggest that CS regulates a broad spectrum of human cellular processes. Full article
(This article belongs to the Special Issue Molecular Advances in Cancer and Cell Metabolism—2nd Edition)
Show Figures

Figure 1

Back to TopTop