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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (293)

Search Parameters:
Keywords = mitochondrial membrane depolarization

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 4979 KB  
Article
Investigating the Potential Role of Capsaicin in Facilitating the Spread of Coxsackievirus B3 via Extracellular Vesicles
by Shruti Chatterjee, Ramina Kordbacheh, Haylee Tilley, Devin Briordy, Richard T. Waldron, William D. Cutts, Jayden Aleman, Alexis Cook, Raeesa Dhanji, Lok-Yin Roy Wong, Stephen J. Pandol, Brandon J. Kim, DeLisa Fairweather and Jon Sin
Int. J. Mol. Sci. 2026, 27(2), 661; https://doi.org/10.3390/ijms27020661 - 9 Jan 2026
Viewed by 146
Abstract
Coxsackievirus B3 (CVB3) is a picornavirus that causes systemic inflammatory diseases including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. We have previously reported that CVB3 induces mitochondrial fission and mitophagy while inhibiting lysosomal degradation by blocking autophagosome-lysosome fusion. This promotes the release of virus-laden mitophagosomes [...] Read more.
Coxsackievirus B3 (CVB3) is a picornavirus that causes systemic inflammatory diseases including myocarditis, pericarditis, pancreatitis, and meningoencephalitis. We have previously reported that CVB3 induces mitochondrial fission and mitophagy while inhibiting lysosomal degradation by blocking autophagosome-lysosome fusion. This promotes the release of virus-laden mitophagosomes from host cells as infectious extracellular vesicles (EVs), enabling non-lytic viral egress. Transient receptor potential vanilloid 1 (TRPV1), a heat and capsaicin-sensitive cation channel, regulates mitochondrial dynamics by inducing mitochondrial membrane depolarization and fission. In this study, we found that TRPV1 activation by capsaicin dramatically enhances CVB3 egress from host cells via EVs. Released EVs revealed increased levels of viral capsid protein VP1, mitochondrial protein TOM70, and fission protein phospho-DRP1. Moreover, these EVs were enriched in heat shock protein HSP70, suggesting its role in facilitating infectious EV release from cells. Furthermore, TRPV1 inhibition with capsazepine and SB-366791 significantly reduced viral infection in vitro. Our in vivo studies also found that SB-366791 significantly mitigates pancreatic damage and reduces viral titers in a mouse model of CVB3 pancreatitis. Given the lack of understanding regarding factors that contribute to diverse clinical manifestations of CVB3, our study highlights capsaicin and TRPV1 as potential exacerbating factors that facilitate CVB3 dissemination via mitophagy-derived EVs. Full article
Show Figures

Figure 1

24 pages, 2905 KB  
Article
Evaluation of Cedrus atlantica Essential Oil: Chemical Composition, Anticancer Activity and Molecular Docking Studies
by Silvia Gruin, Octavian Crețu, Alexandra Mioc, Marius Mioc, Alexandra Prodea, Elisabeta Atyim, Alexandra Teodora Lukinich-Gruia, Maria-Alexandra Pricop, Armand Gogulescu and Codruța Șoica
Molecules 2026, 31(1), 46; https://doi.org/10.3390/molecules31010046 - 22 Dec 2025
Viewed by 497
Abstract
Due to their high content of bioactive compounds with anticancer properties, essential oils (EO) are increasingly viewed as valuable therapeutic strategies in oncology. The aim of this study was to evaluate the chemical composition and anticancer activity of Cedrus atlantica EO (CAEO) and [...] Read more.
Due to their high content of bioactive compounds with anticancer properties, essential oils (EO) are increasingly viewed as valuable therapeutic strategies in oncology. The aim of this study was to evaluate the chemical composition and anticancer activity of Cedrus atlantica EO (CAEO) and its PEG-400 and Tween 20 formulations. The gas-chromatography (GC) analysis revealed a sesquiterpene-rich profile, with β-himachalene (39.32%) as the major constituent, followed by α-Himachalene (16.76%) and γ-Himachalene (12.92%). The cytotoxicity studies, performed using Alamar Blue assay on normal HaCaT human keratinocytes and A375 human melanoma and HT-29 colorectal carcinoma cell lines, revealed that CAEO displayed minimal toxicity on HaCaT cells, while significantly reducing A735 and HT-29 cell viability, at any of the concentrations tested. The PEG- and Tween-based formulations of CAEO exhibited the same effect on cell viability as the simple water dispersion of CAEO. The immunofluorescence-based examination of cellular morphology suggested that CAEO induces apoptosis in both cancer cell lines: A375 and HT-29; this apoptosis-related mechanism was further supported by the caspase-3/7 assay, which revealed a significant increase in caspase-3/7 activity after CAEO treatment. To further investigate the underlying mechanism, the JC-1 staining and high-resolution respirometry assays demonstrated that CAEO induces mitochondrial membrane depolarization and reduced mitochondrial active respiration (OXPHOS). Molecular docking further indicated that isoledene and β-himachalene exhibit the highest predicted affinity for PI3Kγ, suggesting a potential involvement of PI3K-related signaling in the pro-apoptotic activity of CAEO. Together, these results suggest that CAEO induces apoptosis through a mitochondria-mediated mechanism. Full article
Show Figures

Graphical abstract

14 pages, 2156 KB  
Article
Effect of Soluble Factors Released from Porcine Freeze-Dried Lung Tissue (FDLT) on Modulation of Cell Growth and EMT Signature in Non-Small Cell Lung Cancer (NSCLC)—A Preliminary In Vitro Study
by Umme Samia, Daniela Omodei, Luisa Amato, Caterina De Rosa, Rosa Camerlingo, Virna Conti, Stefano Grolli, Orlando Ferroni, Adriano Piattelli, Giovanni N. Roviello, Carminia Maria Della Corte, Viviana De Rosa, Maria Cristina Curia and Francesca Iommelli
Int. J. Mol. Sci. 2025, 26(23), 11743; https://doi.org/10.3390/ijms262311743 - 4 Dec 2025
Viewed by 447
Abstract
Lung cancer remains one of the leading causes of cancer-related mortality worldwide, with therapeutic efficacy often hindered by the development of multidrug resistance. Consequently, alternative strategies to slow down tumor progression warrant rigorous investigation. Bioactive molecules derived from tissues and organs have shown [...] Read more.
Lung cancer remains one of the leading causes of cancer-related mortality worldwide, with therapeutic efficacy often hindered by the development of multidrug resistance. Consequently, alternative strategies to slow down tumor progression warrant rigorous investigation. Bioactive molecules derived from tissues and organs have shown potential therapeutic properties for several diseases. We investigated the biological role of soluble bioactive factors derived from lyophilized porcine freeze-dried lung tissue (FDLT), as they may contain tumor-suppressing components involved in the progression of non-small cell lung cancer (NSCLC). NSCLC H1975 and PC9 cell lines were treated with FDLT at concentrations of 0.25 mg/mL and 0.5 mg/mL. Cell cycle analysis and mitochondrial membrane potential (MMP) assays were performed to assess cell proliferation and cell death activation. In parallel, epithelial–mesenchymal transition (EMT) markers were detected by qRT-PCR. Our findings showed that FDLT treatment reduced the viability of H1975 and PC9 cells in a dose-dependent manner, along with significant suppression of cell proliferation and colony formation. Moreover, FDLT treatment altered the cell cycle phases and determined a concomitant reduction of cyclin D1 levels as well as induction of mitochondria depolarization by suppressing MMP. Finally, qRT-PCR revealed significant downregulation of EMT-related genes vimentin and N-cadherin, along with the EMT transcription factor Twist. These findings highlight soluble FDLT-derived biomolecules as a potential tool to design alternative treatment strategies for NSCLC. Full article
Show Figures

Figure 1

15 pages, 3408 KB  
Article
Therapeutic Effects of Cephalotaxus harringtonia Leaf Extract on Hepatocellular Carcinoma via Regulation of the Intrinsic Apoptosis Pathway and Cell Cycle
by Dae-Han Park, Sonny C. Ramos, Hyun Bo Sim, Ju-Bin Lee, Ho-Yeol Jang, Beom-Gyun Jeong, Kyung-Wuk Park, Kyung-Yun Kang and Jong-Jin Kim
Curr. Issues Mol. Biol. 2025, 47(12), 994; https://doi.org/10.3390/cimb47120994 - 27 Nov 2025
Cited by 1 | Viewed by 440
Abstract
Apoptosis induction in tumor cells is a fundamental therapeutic approach in cancer treatment, with growing interest in plant-derived compounds that offer potent efficacy and reduced toxicity. Cephalotaxus harringtonia, traditionally used in East Asian medicine, contains several bioactive constituents, including homoharringtonine (HHT) and [...] Read more.
Apoptosis induction in tumor cells is a fundamental therapeutic approach in cancer treatment, with growing interest in plant-derived compounds that offer potent efficacy and reduced toxicity. Cephalotaxus harringtonia, traditionally used in East Asian medicine, contains several bioactive constituents, including homoharringtonine (HHT) and quercetin 3-β-D-glucoside (Q3G), which are known for their anticancer properties. This study investigated the anticancer effects of C. harringtonia leaf extract (CHLE) and its two major compounds, quercetin 3-β-D-glucoside (Q3G) and HHT, against human liver cancer cell lines (HepG2). CHLE exhibited selective cytotoxicity and apoptosis specifically in HepG2 cells while showing minimal toxicity toward normal kidney cells (HK-2). Mechanistic analyses revealed that CHLE induced apoptosis through a mitochondria-mediated intrinsic pathway, characterized by increased reactive oxygen species production, mitochondrial membrane depolarization, and BAX upregulation. These findings demonstrate that C. harringtonia leaf extract possesses potent, selective anticancer activity and may serve as a promising natural candidate for the prevention and therapeutic management of liver cancer. Full article
Show Figures

Figure 1

26 pages, 27449 KB  
Article
Ferritin Mitochondrial (FTMT)-Driven Mitochondrial Ferroptosis in Vascular Smooth Muscle Cells: A Role of NCOA4 in Atherosclerosis Pathogenesis and Modulation by Gualou–Xiebai
by Li Zhu, Jun Gao, Zijian Liu, An Zhou and Hongfei Wu
Nutrients 2025, 17(23), 3713; https://doi.org/10.3390/nu17233713 - 26 Nov 2025
Viewed by 738
Abstract
Background/Objectives: Atherosclerosis (AS)-related cardiovascular diseases are a major global health threat, with vascular smooth muscle cells (VSMCs) phenotypic switching, abnormal proliferation, and migration as key progression drivers. Nuclear receptor coactivator 4 (NCOA4), a core ferritinophagy mediator overexpressed in AS plaques, may promote [...] Read more.
Background/Objectives: Atherosclerosis (AS)-related cardiovascular diseases are a major global health threat, with vascular smooth muscle cells (VSMCs) phenotypic switching, abnormal proliferation, and migration as key progression drivers. Nuclear receptor coactivator 4 (NCOA4), a core ferritinophagy mediator overexpressed in AS plaques, may promote VSMCs ferroptosis by perturbing mitochondrial iron metabolism and ROS homeostasis, but precise mechanisms remain unclear. The classic Chinese herbal combination “Gualou-Xiebai” (GLXB) has anti-AS effects, yet how it modulates NCOA4-mediated ferroptosis to inhibit VSMCs’ functions is unknown. This study addresses this gap to advance GLXB’s therapeutic potential and identify AS targets. Methods: An AS model was established in ApoE−/− mice by 12-week high-fat diet feeding, with model validation confirmed via ultrasound monitoring and H&E staining. NCOA4 was genetically modulated (knockdown and overexpression) to assess its role in plaque formation and lipid deposition using H&E staining, aortic imaging, immunofluorescence, and Western blotting. In vitro, VSMCs were stimulated with ox-LDL to induce proliferation and migration. NCOA4 was silenced using siRNA to examine associated ferroptosis levels and molecular mechanisms. Protein interactions between NCOA4 and the mitochondrial iron storage protein FTMT were evaluated by Co-IP and GST pull-down assays, while mitochondrial ROS (mitoROS) levels were measured to explore functional relationships. The extent of ferroptosis and the underlying regulatory mechanisms were assessed following treatment with GLXB-containing serum or transfection with small interfering RNA targeting LOX-1 (si-LOX-1). Results: NCOA4 knockdown reduced aortic lipid deposition, plaque burden, VSMC proliferation/migration, and mitochondrial ferroptosis. NCOA4 bound and suppressed FTMT, inducing mitochondrial iron overload, ROS accumulation, membrane depolarization, and ferroptosis. Combining NCOA4 silencing with FTMT inhibition elevated mitoROS, confirming the axis’s role in iron homeostasis. GLXB attenuated VSMCs dysregulation in vivo and in vitro, an effect abrogated by LOX-1 overexpression. Conclusions: NCOA4 promotes AS by binding FTMT, disrupting mitochondrial iron homeostasis, and triggering VSMCs ferroptosis. GLXB inhibits LOX-1-mediated NCOA4 expression, mitigating ferroptosis and VSMCs dysregulation, supporting its potential as a targeted anti-AS therapy. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

16 pages, 1555 KB  
Article
Lipopolysaccharide Potentiates Platelet Aggregation in Association with Apoptosis Through a Novel TLR4–Bax/Bcl-2-Mitochondrial Dysfunction Axis in Humans
by Chun-Chao Chen, Chih-Wei Hsia, Wei-Chieh Huang, Chao-Chien Chang, Arief Gunawan Darmanto and Joen-Rong Sheu
Biomolecules 2025, 15(12), 1638; https://doi.org/10.3390/biom15121638 - 21 Nov 2025
Viewed by 2460
Abstract
Platelets are anucleate cells whose dysregulation contributes to thrombocytopenia during sepsis. Thrombocytopenia is an early complication of Gram-negative infection, in which lipopolysaccharide (LPS) serves as a principal mediator; however, its precise contribution remains unclear. In this study, LPS, at concentration 10 µg/mL, did [...] Read more.
Platelets are anucleate cells whose dysregulation contributes to thrombocytopenia during sepsis. Thrombocytopenia is an early complication of Gram-negative infection, in which lipopolysaccharide (LPS) serves as a principal mediator; however, its precise contribution remains unclear. In this study, LPS, at concentration 10 µg/mL, did not induce human platelet aggregation but significantly potentiated low-dose collagen (0.5 μg/mL)-induced aggregation, ATP release, intracellular calcium levels ([Ca2+]i) and P-selectin expression. Scanning electron microscopy revealed that either collagen or LPS activated filopodia elongation in human platelets, whereas LPS combined with collagen further activated the phenotype of platelet activation (lamellipodia formation). Beyond these activation responses, LPS also increased TLR4 expression and triggered hallmark apoptotic events, including mitochondrial depolarization, Bax expression, caspase-8 and caspase-3 activation, and phosphatidylserine exposure, concomitant with downregulation of Bcl-2. Moreover, LPS-induced apoptotic platelets displayed ultrastructural changes, characterized by membrane blebbing and filopodia loss. Thus, these findings present the first evidence that LPS enhances platelet aggregation in association with apoptosis through the TLR4–Bax/Bcl-2–mitochondrial dysfunction–caspase-8/3 activation signaling pathway, providing mechanistic insight into sepsis-associated thrombocytopenia. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms in Anti-Thrombosis)
Show Figures

Figure 1

16 pages, 3941 KB  
Article
Bis-Oxadiazole Assemblies as NO-Releasing Anticancer Agents
by Egor M. Matnurov, Irina A. Stebletsova, Alexander A. Larin, Jemma Arakelyan, Ivan V. Ananyev, Artem L. Gushchin, Leonid L. Fershtat and Maria V. Babak
Pharmaceutics 2025, 17(11), 1494; https://doi.org/10.3390/pharmaceutics17111494 - 19 Nov 2025
Viewed by 924
Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive, asbestos-associated cancer characterized by dysregulated nitric oxide (NO) signaling and increased NO levels that facilitate tumor progression. Paradoxically, this aberrant NO environment creates a therapeutic vulnerability that can be exploited by NO-donor prodrugs, which [...] Read more.
Background: Malignant pleural mesothelioma (MPM) is an aggressive, asbestos-associated cancer characterized by dysregulated nitric oxide (NO) signaling and increased NO levels that facilitate tumor progression. Paradoxically, this aberrant NO environment creates a therapeutic vulnerability that can be exploited by NO-donor prodrugs, which overwhelm cellular defenses with cytotoxic concentrations of NO, inducing nitrosative stress and apoptosis. Within this framework, oxadiazole-based scaffolds have emerged as a promising platform for prodrug development owing to their versatile chemistry and potential as novel NO donors or synergistic agents. In our previous studies, we developed several series of hybrid architectures incorporating 1,2,5-oxadiazole 2-oxide (furoxan) and 1,2,4-oxadiazole scaffolds, producing compounds with diverse and tunable NO-donor activities. We further observed that the cytotoxicity of these hybrids was significantly influenced by the substituents introduced at position 3 of the furoxan ring. Methods: We designed and synthesized a series of bis(1,2,4-oxadiazolyl)furoxans to systematically investigate their NO-donating capacity, cytotoxicity against MPM cell lines, selectivity over healthy lung fibroblasts, and underlying anticancer mechanisms. Results: The bis(1,2,4-oxadiazolyl)furoxans exhibited lower overall cytotoxicity but significantly higher selectivity compared with previously studied 3-cyano-4-(1,2,4-oxadiazolyl)furoxans. Their NO-releasing properties showed a strong correlation with their ability to induce mitochondrial damage, as evidenced by membrane depolarization. Moreover, the incorporation of specific substituents, such as a furan ring, on the 1,2,4-oxadiazole moiety introduced an additional mechanism of action through the induction of reactive oxygen species. Conclusions: Analysis of cancer cell death confirmed that these compounds acted through a multimodal mechanism dependent on both NO release and the specific substituents on the 1,2,4-oxadiazole moiety. Full article
(This article belongs to the Special Issue Prodrug Applications for Targeted Cancer Therapy)
Show Figures

Graphical abstract

18 pages, 2398 KB  
Article
Extended Preservation of Heart Grafts: LYPS Solution Maintains Cardiac Function During 20-Hour Static Cold Storage
by Marie Védère, Evan Faure, Christophe Chouabe, Lionel Augeul, Ninon Cadot-Jet, Georges Christé, Yanis Charouit, Mégane Lo Grasso, Alexandre Ravon, Régine Cartier, Gabriel Bidaux, René Ferrera, Hala Guedouari and Delphine Baetz
Int. J. Mol. Sci. 2025, 26(22), 11170; https://doi.org/10.3390/ijms262211170 - 19 Nov 2025
Viewed by 508
Abstract
Heart transplantation is severely limited by the shortage of suitable donor grafts, partly due to myocardial vulnerability to ischemia–reperfusion injury and the lack of standardized preservation strategies. Current solutions only partially maintain myocardial viability, compromising post-transplant function. To address this issue, we made [...] Read more.
Heart transplantation is severely limited by the shortage of suitable donor grafts, partly due to myocardial vulnerability to ischemia–reperfusion injury and the lack of standardized preservation strategies. Current solutions only partially maintain myocardial viability, compromising post-transplant function. To address this issue, we made further improvements to our preservation solution, LYPS (Lyon Preservation Solution), based on mitochondrial metabolic activation and the limitation of membrane depolarization. We first evaluated commonly used extracellular solutions (Celsior and St. Thomas (ST)) on cardiac cell lines (H9C2) exposed to 20 h of cold (4 °C) simulated ischemia followed by 2 h of simulated reperfusion. In parallel, the same three solutions were compared in isolated pig hearts subjected to 20 h of cold static storage followed by reperfusion, with a group directly reperfused with blood at 37 °C serving as the control. Heart function was assessed using a non-working heart preparation, while mitochondrial functions and electrophysiological analysis were evaluated via biopsies and isolated cardiomyocytes. LYPS provided superior protection against cell death and mitochondrial membrane potential loss in vitro, outperformed ST in preserving mitochondrial function, and limited troponin I release by the heart. During reperfusion, LYPS-treated hearts showed improved functional recovery and contractility and better rhythmicity with almost no defibrillation requirements. These effects may involve the modulation of the repolarizing IK1 current. Overall, LYPS effectively preserves myocardial viability and function, representing a promising strategy to enhance graft quality during long-term cold preservation, even through using cold static storage. Full article
(This article belongs to the Special Issue Molecular Insights into Transplantation and Machine Perfusion)
Show Figures

Figure 1

20 pages, 9854 KB  
Article
In Vitro and Ultrastructural Evaluation of the Cytotoxic and Antileishmanial Activities of Thiosemicarbazone Compounds Against Promastigotes and Axenic Amastigotes of Leishmania infantum
by Janderson Weydson Lopes Menezes da Silva, Andréa Regina Alves da Rocha Diniz, Alberon Ribeiro de Araújo, Gabriel Gazzoni Araújo Gonçalves, Dyana Leal Veras, Marton Kaique de Andrade Cavalcante, Jana Messias Sandes, Iranildo José da Cruz Filho, Diego Santa Clara Marques, Maria do Carmo Alves de Lima, Ana Paula Sampaio Feitosa, Luiz Carlos Alves and Fábio André Brayner
Trop. Med. Infect. Dis. 2025, 10(11), 325; https://doi.org/10.3390/tropicalmed10110325 - 19 Nov 2025
Viewed by 548
Abstract
Leishmaniasis remains a global health challenge, and the search for effective and selective therapeutic agents is crucial. This study evaluated the in vitro antileishmanial and cytotoxic activities of thiosemicarbazone compounds (LT-70, LT-73, LT-75, and LT-89) against Leishmania infantum promastigote and axenic amastigote forms. [...] Read more.
Leishmaniasis remains a global health challenge, and the search for effective and selective therapeutic agents is crucial. This study evaluated the in vitro antileishmanial and cytotoxic activities of thiosemicarbazone compounds (LT-70, LT-73, LT-75, and LT-89) against Leishmania infantum promastigote and axenic amastigote forms. The compounds demonstrated strong leishmanicidal activity, with IC50 values ranging from 10.5 to 14 µM. At the lowest tested concentration (20 µM) the compounds produced percent inhibitions of 100% (LT-70), 100% (LT-73), 100% (LT-75) and 100% (LT-89). Cytotoxicity assays on J774.A1 macrophages revealed CC50 values from 60 µM to >75 µM, with LT-73 and LT-75 showing low toxicity (CC50 > 75µM). Selectivity index (SI) ranged from 7.1 for LT-75 and 5.8 for LT-73, indicating potential for further development. Ultrastructural analysis by SEM and TEM revealed cellular and organelle damage, including membrane rupture and mitochondrial swelling, especially after LT-73 and LT-75 treatment. Immunomodulatory assays indicated induction of TNF-α and IFN-γ production, with significant IL-6 reduction. Flow cytometry data suggest mitochondrial dysfunction and apoptosis-like features, particularly for LT-73. Membrane potential assays suggested mitochondrial depolarization by LT-73. LT thiosemicarbazone derivatives present specific structural modifications that enhance antileishmanial selectivity and reveal a dual mechanism of action combining mitochondrial dysfunction and immunomodulatory effects. These findings support the potential of thiosemicarbazone derivatives as promising antileishmanial agents with selective cytotoxicity and immunomodulatory effects. Full article
Show Figures

Figure 1

18 pages, 2610 KB  
Article
Exploring the Biochemical Mechanism Beyond the Cytotoxic Activity of Sesquiterpene Lactones from Sicilian Accession of Laserpitium siler Subsp. siculum (Spreng.) Thell
by Alessandro Vaglica, Antonella Maggio, Chiara Occhipinti, Natale Badalamenti, Marianna Lauricella, Maurizio Bruno and Antonella D’Anneo
Plants 2025, 14(21), 3289; https://doi.org/10.3390/plants14213289 - 28 Oct 2025
Viewed by 645
Abstract
Laserpitium siler subsp. siculum (Apiaceae) is a Mediterranean plant with a long history of traditional medicinal use. In this study, the chemical composition and anticancer potential of three novel (and one new to the genus) sesquiterpene lactones (SLs) isolated from its roots were [...] Read more.
Laserpitium siler subsp. siculum (Apiaceae) is a Mediterranean plant with a long history of traditional medicinal use. In this study, the chemical composition and anticancer potential of three novel (and one new to the genus) sesquiterpene lactones (SLs) isolated from its roots were investigated. The structural characterization, carried out through NMR and HPLC-MS analyses, identified unique guaianolide-type lactones. The biological activity of these compounds was evaluated in vitro using MDA-MB-231 cells, a triple-negative breast cancer (TNBC) cell line. Cell viability assays demonstrated that all SLs tested reduced TNBC cell viability in a dose- and time-dependent manner, with SL-1 exhibiting the highest cytotoxicity. Light microscopy analyses and acridine orange/ethidium bromide staining confirmed the induction of apoptotic cell death, further supported by Western blot analyses showing caspase-3 activation and PARP-1 cleavage. Additional experiments indicated that SL-1 induced oxidative stress, as evidenced by increased ROS production and upregulation of the levels of the antioxidant enzymes MnSOD and HO-1. Moreover, JC-1 staining and Western blot analyses revealed mitochondrial membrane depolarization as well as a significant reduction in VDAC-1 expression, suggesting mitochondrial dysfunction as a key event in the cytotoxic mechanism. These findings highlight L. siler subsp. siculum as a promising source of bioactive compounds with anticancer potential. The ability of its sesquiterpene lactones to induce oxidative stress and mitochondrial impairment provides new insights into their mode of action, supporting further research into their therapeutic applications for TNBC treatment. Full article
(This article belongs to the Section Phytochemistry)
Show Figures

Figure 1

32 pages, 9810 KB  
Article
Silver(I) Complexes Bearing S-Alkyl Thiosalicylic Acid Derivatives: DNA/BSA Binding and Antitumor Activity In Vitro and In Vivo
by Jovana Marinković, Milena Jurišević, Marina Jovanović, Miloš Milosavljević, Ivan Jovanović, Snežana Jovanović Stević, Marina Vesović, Miloš Nikolić, Nikola Nedeljković, Ana Živanović, Dušan Tomović, Andriana Bukonjić, Gordana Radić and Nevena Gajović
Pharmaceutics 2025, 17(10), 1340; https://doi.org/10.3390/pharmaceutics17101340 - 16 Oct 2025
Viewed by 815
Abstract
Background/Objectives: In recent years, silver complexes have shown strong antibacterial, antifungal, and antitumor activity with high selectivity toward cancer cells. Their cytotoxic effects are mainly linked to apoptosis induction, DNA damage, and enzyme inhibition, while the antitumor activity of silver(I) complexes with S-alkyl [...] Read more.
Background/Objectives: In recent years, silver complexes have shown strong antibacterial, antifungal, and antitumor activity with high selectivity toward cancer cells. Their cytotoxic effects are mainly linked to apoptosis induction, DNA damage, and enzyme inhibition, while the antitumor activity of silver(I) complexes with S-alkyl thiosalicylic acid derivatives remains unexplored. Methods: Silver(I) complexes with S-alkyl derivatives of thiosalicylic acid (C1C5) were obtained through the direct reaction of silver(I) nitrate, the corresponding ligand of thiosalicylic acid, and a sodium hydroxide solution. The interactions between the complexes and CT-DNA/BSA were studied using UV-Vis, fluorescence spectroscopy, and molecular docking studies. The cytotoxic capacity of the newly synthesized complexes was assessed by an MTT assay. Results: Complexes C1C5 exhibited strong cytotoxicity against murine and human breast (4T1, MDA-MB-468), colon (CT26, HCT116), and lung (LLC1, A549) cancer cell lines. The C3 complex significantly diminished tumor progression in an orthotropic mammary carcinoma model while demonstrating good systemic tolerance. Conclusions: The tested complex C3 triggered apoptosis in 4T1 cells by altering the delicate balance between pro- and anti-apoptotic Bcl-2 family members, increasing reactive oxygen species (ROS) levels, and reducing mitochondrial membrane depolarization. Moreover, the C3 arrested the 4T1 cell cycle in G0/G1 phase, decreasing the expression of cyclin D3 and increasing the expression of p16, p21, and p27. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

17 pages, 1229 KB  
Article
Pro-Apoptotic and Cytotoxic Effects of Melittin on HL-60 Acute Promyelocytic Leukemia Cells: Implications for Retinoid-Independent Therapy
by Maksymilian Stela, Michał Ceremuga, Natalia Cichon, Tomasz Poplawski, Marcin Podogrocki, Leslaw Gorniak and Michał Bijak
Molecules 2025, 30(20), 4093; https://doi.org/10.3390/molecules30204093 - 15 Oct 2025
Viewed by 1008
Abstract
Background: Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia driven by the PML/RARα fusion protein. Standard treatment with all-trans retinoic acid (ATRA) combined with chemotherapy is effective, but resistance and adverse effects remain significant challenges. Melittin, the primary peptide component [...] Read more.
Background: Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia driven by the PML/RARα fusion protein. Standard treatment with all-trans retinoic acid (ATRA) combined with chemotherapy is effective, but resistance and adverse effects remain significant challenges. Melittin, the primary peptide component of bee venom, has demonstrated potent anticancer activity across multiple leukemia subtypes through mitochondrial-dependent mechanisms. Building upon this established evidence, we investigated melittin’s therapeutic potential in APL to address the specific clinical challenge of ATRA resistance. Methods: The cytotoxic and pro-apoptotic effects of melittin were studied on the human APL cell line HL-60. Cell viability was assessed using MTT and trypan blue assays. Mitochondrial membrane potential (MMP) was measured with JC-1 staining. Apoptosis was quantified using Annexin V/propidium iodide flow cytometry, caspase-3/7 activity assays, and real-time PCR analysis of apoptosis-related genes (BCL-2, BAX, APAF-1, CASP-3, CASP-8, CASP-9). Results: Melittin reduced HL-60 cell viability in a dose- and time-dependent manner, with significant decreases after 24 and 48 h. MMP analysis revealed mitochondrial depolarization, and Annexin V staining confirmed the induction of apoptosis. Caspase-3/7 activity increased markedly, supporting activation of the intrinsic apoptotic pathway. Gene expression profiling revealed downregulation of the anti-apoptotic BCL-2 and upregulation of the pro-apoptotic BAX, APAF1, and CASP3. At the same time, CASP8 and CASP9 showed no significant changes, suggesting a predominant involvement of the intrinsic pathway. Conclusions: These findings confirm and extend established evidence by demonstrating that melittin’s mitochondrial apoptotic mechanism is consistently active in promyelocytic HL-60 model (PML/RARα-negative). This retinoid-independent mechanism suggests potential therapeutic utility for ATRA-resistant cases or as a complementary strategy in APL treatment. However, selectivity validation in non-cancerous hematopoietic cells represents an important future research priority. Full article
Show Figures

Figure 1

24 pages, 9190 KB  
Article
Targeting Glycolysis with 2-Deoxy-D-Glucose and Lysosomal Integrity with L-Leucyl-L-Leucine Methyl Ester as Antimelanoma Strategy
by Milica Kosic, Mihajlo Bosnjak, Milos Mandic, Ljubica Vucicevic, Maja Misirkic Marjanovic, Sofie Espersen Poulsen and Ljubica Harhaji-Trajkovic
Pharmaceutics 2025, 17(10), 1312; https://doi.org/10.3390/pharmaceutics17101312 - 9 Oct 2025
Viewed by 997
Abstract
Background/Objectives: Melanoma cells enhance glycolysis and expand lysosomes to support energy metabolism, proliferation, and metastasis. However, lysosomal membrane permeabilization (LMP) causes cathepsin leakage into cytosol triggering cytotoxicity. This study investigated the antimelanoma effect of 2-deoxy-D-glucose (2DG), an inhibitor of glycolytic enzyme hexokinase-2, [...] Read more.
Background/Objectives: Melanoma cells enhance glycolysis and expand lysosomes to support energy metabolism, proliferation, and metastasis. However, lysosomal membrane permeabilization (LMP) causes cathepsin leakage into cytosol triggering cytotoxicity. This study investigated the antimelanoma effect of 2-deoxy-D-glucose (2DG), an inhibitor of glycolytic enzyme hexokinase-2, in combination with cathepsin C-dependent LMP inducer L-leucyl-L-leucine methyl ester (LLOMe) and cathepsin C-independent LMP-inducers mefloquine and siramesine. Methods: The viability of A375 and B16 melanoma cells and primary fibroblasts was measured by crystal violet. Apoptosis, necrosis, and LMP were assessed by flow cytometry; caspase activation, mitochondrial depolarization, superoxide production, and energy metabolism were analyzed by fluorimetry, and expression of cathepsins and hexokinase-2 was evaluated by immunoblot. Appropriate inhibitors, antioxidant, and energy boosters were used to confirm cell death type and mechanism. Results: LLOMe triggered LMP, mitochondrial depolarization, and mitochondrial superoxide production, while suppressing oxidative phosphorylation. 2DG suppressed glycolysis and, together with LLOMe, synergized in ATP depletion, caspase activation, and mixed apoptosis and necrosis in A375 cells. Inhibitors of lysosomal acidification, cysteine cathepsins, and caspases, as well as antioxidant and energy boosters, reduced 2DG+LLOMe-induced toxicity. Cathepsins B, C, and D were lower, while hexokinase-2 was higher in A375 cells than fibroblasts. Accordingly, 2DG exhibited lower while LLOMe exhibited higher toxicity against fibroblasts than A375 and B16 cells. However, mefloquine and siramesine induced stronger LMP in A375 cells than in fibroblasts and showed melanoma-selective toxicity when combined with 2DG. Conclusions: 2DG-mediated glycolysis inhibition in combination with lysosomal destabilization induced by mefloquine and siramesine, but not with non-selectively toxic LLOMe, may be promising antimelanoma strategy. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

15 pages, 4644 KB  
Article
Cardioprotective Effects of Simvastatin in Doxorubicin-Induced Acute Cardiomyocyte Injury
by Roberta Vitale, Mariangela Mazzone, Maria Carmela Di Marcantonio, Stefania Marzocco, Gabriella Mincione and Ada Popolo
Int. J. Mol. Sci. 2025, 26(19), 9440; https://doi.org/10.3390/ijms26199440 - 26 Sep 2025
Cited by 1 | Viewed by 789
Abstract
Oxidative stress and mitochondrial dysfunction play a key role in the early stage of Doxorubicin (Doxo)-induced cardiotoxicity. Our study investigated the potential cardioprotective role of Simvastatin (Sim), widely known for its antioxidant properties, in an in vitro model of Doxo-induced acute cardiotoxicity. Human [...] Read more.
Oxidative stress and mitochondrial dysfunction play a key role in the early stage of Doxorubicin (Doxo)-induced cardiotoxicity. Our study investigated the potential cardioprotective role of Simvastatin (Sim), widely known for its antioxidant properties, in an in vitro model of Doxo-induced acute cardiotoxicity. Human Cardiomyocytes (HCMs) were treated with Sim (10 µM, 4 h) and then co-exposed to Doxo (1 µM) and Sim for 20 h. Our data showed that Sim co-treatment significantly (p < 0.05) reduced both cytosolic and mitochondrial Doxo-induced reactive oxygen species overproduction. In Sim co-treated cells, significant reductions in nuclear factor erythroid 2-related factor 2 (Nrf2) gene expression (p < 0.01) and catalase (CAT), heme-oxygenase 1 (HO-1), and superoxide dismutase 2 (SOD2) levels (p < 0.05) compared to Doxo-treated cells were also demonstrated, suggesting a decreased need for compensatory antioxidant defense responses. Moreover, significant reductions in Doxo-induced mitochondrial calcium overload, mitochondrial membrane depolarization (p < 0.005), and apoptosis (p < 0.005) confirmed the protective effects of Sim co-treatment on cardiomyocytes. These data confirm that Sim could be a valuable therapeutic strategy for reducing Doxo-induced HCM damage, preventing the development of dilated cardiomyopathy and long-term heart damage, which are the main limitations of anthracycline use. Finally, real-time PCR analysis revealed that Sim co-treatment significantly reduced (p < 0.001) the Doxo-induced overexpression of MAP4K4, a mitogen-activated protein kinase kinase kinase kinase-4 (MAP4K4) involved in oxidative stress-induced cell death, thus suggesting the involvement of other molecular mechanisms in Sim-mediated cardioprotection. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

16 pages, 1890 KB  
Article
Cellular Mechanisms of Photobiomodulation in Relation to HeLa Kyoto Tumor Cells Exposed to Ionizing Radiation
by Anna V. Maslennikova, Artem O. Belotelov, Elena I. Cherkasova, Vladimir I. Yusupov, Ulyana A. Kononova, Natalia Yu. Shilyagina and Dmitry V. Skamnitsky
Int. J. Mol. Sci. 2025, 26(18), 9197; https://doi.org/10.3390/ijms26189197 - 20 Sep 2025
Viewed by 911
Abstract
During the clinical use of photobiomodulation (PBM) to manage radiotherapy-induced side effects, tumor tissue may be exposed to low-intensity laser light. Therefore, it is necessary to evaluate potential unintended PBM stimulation of tumor cells when PBM combines with ionizing radiation (IR). We investigated [...] Read more.
During the clinical use of photobiomodulation (PBM) to manage radiotherapy-induced side effects, tumor tissue may be exposed to low-intensity laser light. Therefore, it is necessary to evaluate potential unintended PBM stimulation of tumor cells when PBM combines with ionizing radiation (IR). We investigated the effects of PBM (0.3 J/cm2) on the cell cycle, mitochondrial potential, and cell death of HeLa Kyoto cells, comparing pre-irradiation (PBM→IR) and post-irradiation (IR→PBM) exposure to 2 Gy, 4 Gy, and 6 Gy of ionizing radiation. PBM prior to IR induced the radiation-induced arrest in the G2/M phase of the cell cycle. PBM after IR resulted in a partial release of cells from the radiation-induced arrest in the G0/G1 phase, along with a decrease in the number of apoptotic cells and cells with depolarized mitochondrial membranes compared to samples treated with IR only. These findings provide a basis for further research into PBM timing to improve radiotherapy outcomes. Full article
Show Figures

Figure 1

Back to TopTop