Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (84)

Search Parameters:
Keywords = anti-glycolysis therapy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 7271 KiB  
Article
ENO1 from Mycoplasma bovis Disrupts Host Glycolysis and Inflammation by Binding ACTB
by Rui-Rui Li, Xiao-Jiao Yu, Jia-Yin Liang, Jin-Liang Sheng, Hui Zhang, Chuang-Fu Chen, Zhong-Chen Ma and Yong Wang
Biomolecules 2025, 15(8), 1107; https://doi.org/10.3390/biom15081107 - 1 Aug 2025
Viewed by 262
Abstract
Mycoplasma bovis is an important pathogen that is associated with respiratory diseases, mastitis, and arthritis in cattle, leading to significant economic losses in the global cattle industry. Most notably in this study, we pioneer the discovery that its secreted effector ENO1 (α-enolase) directly [...] Read more.
Mycoplasma bovis is an important pathogen that is associated with respiratory diseases, mastitis, and arthritis in cattle, leading to significant economic losses in the global cattle industry. Most notably in this study, we pioneer the discovery that its secreted effector ENO1 (α-enolase) directly targets host cytoskeletal proteins for metabolic–immune regulation. Using an innovative GST pull-down/mass spectrometry approach, we made the seminal discovery of β-actin (ACTB) as the primary host target of ENO1—the first reported bacterial effector–cytoskeleton interaction mediating metabolic reprogramming. ENO1–ACTB binding depends on a hydrogen bond network involving ACTB’s 117Glu and 372Arg residues. This interaction triggers (1) glycolytic activation via Glut1 upregulation, establishing Warburg effect characteristics (lactic acid accumulation/ATP inhibition), and (2) ROS-mediated activation of dual inflammatory axes (HIF-1α/IL-1β and IL-6/TNF-α). This work establishes three groundbreaking concepts: (1) the first evidence of a pathogen effector hijacking host ACTB for metabolic manipulation, (2) a novel ‘glycolysis–ACTB–ROS-inflammation’ axis, and (3) the first demonstration of bacterial proteins coordinating a Warburg effect with cytokine storms. These findings provide new targets for anti-infection therapies against Mycoplasma bovis. Full article
(This article belongs to the Section Biomacromolecules: Proteins, Nucleic Acids and Carbohydrates)
Show Figures

Figure 1

15 pages, 1151 KiB  
Review
Mitochondrial Dysfunction and Glycolytic Shift in the Tumor Microenvironment: Impact on Paclitaxel Efficacy in Cancer Therapy
by Tanvi Premchandani, Jayshree Taksande, Amol Tatode, Sameer Sheikh, Mohammad Qutub, Ujban Md Hussain, Rahmuddin Khan and Milind Umekar
Clin. Bioenerg. 2025, 1(1), 5; https://doi.org/10.3390/clinbioenerg1010005 - 9 Jul 2025
Viewed by 344
Abstract
Tumor cells often exhibit mitochondrial dysfunction and a pronounced glycolytic shift (the “Warburg effect”) that alters the tumor microenvironment. These metabolic changes, including mitochondrial DNA mutations and impaired oxidative phosphorylation, confer survival advantages and can reduce sensitivity to chemotherapeutics such as paclitaxel. In [...] Read more.
Tumor cells often exhibit mitochondrial dysfunction and a pronounced glycolytic shift (the “Warburg effect”) that alters the tumor microenvironment. These metabolic changes, including mitochondrial DNA mutations and impaired oxidative phosphorylation, confer survival advantages and can reduce sensitivity to chemotherapeutics such as paclitaxel. In hypoxic environments, cancer cells upregulate glycolysis via HIF-1α, consequently lowering the extracellular pH through lactate secretion, which is associated with resistance to paclitaxel. Likewise, cancer-associated fibroblasts and immune cells undergo metabolic reprogramming in the tumor microenvironment. Glycolytic CAFs produce lactate and pyruvate that fuel tumor cells, reinforcing drug resistance, and tumor-driven polarization of macrophages toward an immunosuppressive M2 phenotype further impairs the anti-tumor response. Here, we review recent findings on how these metabolic adaptations attenuate paclitaxel efficacy and discuss strategies to overcome resistance. We highlight 15 key studies that reported cancer types, metabolic alterations, molecular targets, and outcomes related to paclitaxel response. Overall, the data suggest that targeting metabolic vulnerabilities, for example, by inhibiting glycolysis (HK2, PGAM1, and PDK) or modulating mitochondrial function, may restore paclitaxel sensitivity. Understanding metabolic crosstalk in the tumor microenvironment provides a basis for combined therapies that improve outcomes in paclitaxel-resistant cancers. Full article
Show Figures

Figure 1

17 pages, 2596 KiB  
Article
Low-Dose Salinomycin Alters Mitochondrial Function and Reprograms Global Metabolism in Burkitt Lymphoma
by Aleksandra Zdanowicz, Oleksandr Ilchenko, Andrzej Ciechanowicz, Haoyu Chi, Marta Struga and Beata Pyrzynska
Int. J. Mol. Sci. 2025, 26(11), 5125; https://doi.org/10.3390/ijms26115125 - 27 May 2025
Viewed by 621
Abstract
Salinomycin (SAL), originally identified for its potent antibacterial properties, has recently garnered attention for its remarkable activity against a variety of cancer types. Beyond its direct cytotoxic effects on cancer cells, SAL can also enhance the efficacy of anti-CD20 immunotherapy in B-cell malignancies, [...] Read more.
Salinomycin (SAL), originally identified for its potent antibacterial properties, has recently garnered attention for its remarkable activity against a variety of cancer types. Beyond its direct cytotoxic effects on cancer cells, SAL can also enhance the efficacy of anti-CD20 immunotherapy in B-cell malignancies, both in vitro and in vivo. Despite these promising findings, the precise molecular mechanisms underlying SAL’s anticancer action remain poorly understood. Here, we demonstrate that even at low concentrations (0.25–0.5 mM), SAL disrupts mitochondrial membrane potential and induces oxidative stress in Burkitt lymphoma. Further investigations uncovered that SAL shifts cellular metabolism from mitochondrial respiration to aerobic glycolysis. Additionally, metabolomic profiling identified SAL-induced arginine depletion as a key metabolic alteration. These findings provide new insights into SAL’s multifaceted mechanisms of action and support its potential as an adjunctive therapy in cancer treatment. Full article
(This article belongs to the Special Issue Oxidative Stress and Mitochondria in Human Diseases)
Show Figures

Figure 1

20 pages, 1380 KiB  
Review
The Overlapping Biology of Sepsis and Cancer and Therapeutic Implications
by Amit Kumar Tripathi and Yogesh Srivastava
Biomedicines 2025, 13(6), 1280; https://doi.org/10.3390/biomedicines13061280 - 23 May 2025
Viewed by 953
Abstract
Sepsis and cancer, though distinct in their clinical manifestations, share profound pathophysiological overlaps that underscore their interconnectedness in disease progression and outcomes. Here we discuss the intricate biological mechanisms linking these two conditions, focusing on the roles of inflammation, immune dysregulation, and metabolic [...] Read more.
Sepsis and cancer, though distinct in their clinical manifestations, share profound pathophysiological overlaps that underscore their interconnectedness in disease progression and outcomes. Here we discuss the intricate biological mechanisms linking these two conditions, focusing on the roles of inflammation, immune dysregulation, and metabolic alterations. In sepsis, an uncontrolled immune response to infection leads to a cytokine storm, tissue damage, and immune paralysis, while cancer exploits chronic inflammation and immunosuppressive pathways to promote tumor growth and metastasis. Both conditions exhibit metabolic reprogramming, such as the Warburg effect in cancer and glycolysis-driven immune cell activation in sepsis, which fuels disease progression and complicates treatment. Sepsis can exacerbate cancer progression by inducing genomic instability, epigenetic modifications, and a pro-tumorigenic microenvironment, while cancer increases susceptibility to sepsis through immunosuppression and treatment-related complications. The shared pathways between sepsis and cancer present unique opportunities for therapeutic intervention, including anti-inflammatory agents, immune checkpoint inhibitors, and metabolic modulators. Anti-inflammatory therapies, such as IL-6 and TNF-α inhibitors, show promise in mitigating inflammation, while immune checkpoint inhibitors like anti-PD-1 and anti-CTLA-4 antibodies are being explored to restore immune function in sepsis and enhance antitumor immunity in cancer. Metabolic modulators, including glycolysis and glutaminolysis inhibitors, target the metabolic reprogramming common to both conditions, though their dual roles in normal and pathological processes necessitate careful consideration. Additionally, antimicrobial peptides (AMPs) represent a versatile therapeutic option with their dual antimicrobial and antitumor properties. In this review, we also highlight the critical need for integrated approaches to understanding and managing the complex interactions between sepsis and cancer. By bridging the gap between sepsis and cancer research, this work aims to inspire interdisciplinary collaboration and advance the development of targeted therapies that address the shared mechanisms driving these devastating diseases. Ultimately, these insights may pave the way for novel diagnostic tools and therapeutic strategies to improve outcomes for patients affected by both conditions. Full article
(This article belongs to the Special Issue Sepsis and Septic Shock: From Molecular Mechanism to Novel Therapies)
Show Figures

Figure 1

19 pages, 5160 KiB  
Article
Synergistic Effects of 2-Deoxyglucose and Diclofenac Sodium on Breast Cancer Cells: A Comparative Evaluation of MDA-231 and MCF7 Cells
by Geofrey Ouma Maloba, Tom Were, Erick Barasa, Nasreldeen Mohamed, Arshi Arshi and Ferenc Gallyas
Int. J. Mol. Sci. 2025, 26(10), 4894; https://doi.org/10.3390/ijms26104894 - 20 May 2025
Viewed by 575
Abstract
Resistance of breast cancers to chemotherapy remains a global challenge to date. Drug combination studies between anti-cancer agents are increasingly becoming therapeutic strategies, geared towards alleviating breast cancers. Previously, 2-deoxyglucose has been shown to target and interrupt glycolysis. Available evidence also suggests that [...] Read more.
Resistance of breast cancers to chemotherapy remains a global challenge to date. Drug combination studies between anti-cancer agents are increasingly becoming therapeutic strategies, geared towards alleviating breast cancers. Previously, 2-deoxyglucose has been shown to target and interrupt glycolysis. Available evidence also suggests that diclofenac, which was originally designed as a pain reliever, could inhibit the proliferation of breast cancer cells. However, the reverse Warburg effect and other metabolic reprogramming mechanisms in breast cancers limit the pharmacological application of both 2-deoxyglucose and diclofenac as mono-therapeutic agents. The present study explores the additive anti-cancer effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells. In this study, MDA-231 and MCF7 cells were treated with 2-deoxyglucose and diclofenac sodium in single and combination doses before being evaluated for viability, cell growth, reactive oxygen species, apoptotic and necrotic phases, and migration abilities. Additionally, immunoblotting of pro-apoptotic proteins, Caspase-3 and Caspase-9, and a hypoxia-inducible factor-1 alpha, was also performed. The results showed that combination treatments of the cells with the drugs exhibited additive anti-cancer effects by limiting proliferation, enhancing cytotoxic reactive oxygen species generation, enhancing apoptosis and necrosis, limiting colony formation and expansion of cells, and inhibiting cell migration. The degrees of cytotoxicity of combined treatments were almost similar in both cell lines, although with minimal differences. Put together, these results reveal the novel synergistic effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells, hence potentially elevating their pharmacological profile in the overall breast cancer therapy. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

16 pages, 1998 KiB  
Article
Nicotinamide Inhibits CD4+ T-Cell Activation and Function
by Lotte Nijhuis, Alejandra Bodelόn, Rianne C. Scholman, Isabelle Houtzager, Lyanne J. P. M. Sijbers, Enric Mocholi, Lucas W. Picavet, Jorg J. A. Calis, Michal Mokry, Sebastiaan J. Vastert and Jorg van Loosdregt
Cells 2025, 14(8), 560; https://doi.org/10.3390/cells14080560 - 8 Apr 2025
Viewed by 983
Abstract
Chronic inflammation and autoimmune diseases are driven, in part, by the activation of (auto)reactive CD4+ T-cells, highlighting their potential as therapeutic targets for these diseases. Nicotinamide (NAM) has demonstrated anti-inflammatory properties in various disease models and has already demonstrated safety in several large [...] Read more.
Chronic inflammation and autoimmune diseases are driven, in part, by the activation of (auto)reactive CD4+ T-cells, highlighting their potential as therapeutic targets for these diseases. Nicotinamide (NAM) has demonstrated anti-inflammatory properties in various disease models and has already demonstrated safety in several large clinical trials in humans. The mechanisms behind these observations, and especially their direct effects on CD4+ T-cells, remain poorly understood. Here, we address this gap by investigating how NAM influences CD4+ T-cell activation and function. We also describe that NAM treatment significantly suppresses CD4+ T-cell activation in vitro, as evidenced by impaired proliferation and reduced expression of surface activation markers. Additionally, NAM treatment resulted in reduced production of pro-inflammatory cytokines, IL-2, IFNy, and IL-17, further highlighting its anti-inflammatory potential. We found that NAM modulates key metabolic processes, including glycolysis and reactive oxygen species (ROS) production—both essential to T-cell activation. Taken together, our findings provide novel mechanistic insight into the regulation of T-cell activation by NAM, suggesting NAM as an attractive candidate for novel therapies targeting immune-related diseases. Full article
(This article belongs to the Special Issue T Cells in Inflammation and Cancer)
Show Figures

Figure 1

20 pages, 3223 KiB  
Article
Emodin and Aloe-Emodin Reduce Cell Growth and Disrupt Metabolic Plasticity in Human Melanoma Cells
by Federica Baldassari, Marcella Bonanomi, Sara Mallia, Matteo Bonas, Elisa Brivio, Tecla Aramini, Danilo Porro and Daniela Gaglio
Nutrients 2025, 17(7), 1113; https://doi.org/10.3390/nu17071113 - 22 Mar 2025
Viewed by 1021
Abstract
Background/Objectives: Melanoma is an aggressive skin cancer with intratumor metabolic heterogeneity, which drives its progression and therapy resistance. Natural anthraquinones, such as emodin and aloe-emodin, exhibit anti-cancer properties, but their effects on metabolic plasticity remain unclear. This study evaluated their impact on [...] Read more.
Background/Objectives: Melanoma is an aggressive skin cancer with intratumor metabolic heterogeneity, which drives its progression and therapy resistance. Natural anthraquinones, such as emodin and aloe-emodin, exhibit anti-cancer properties, but their effects on metabolic plasticity remain unclear. This study evaluated their impact on proliferation and metabolic pathways in heterogenous melanoma human cell lines. Methods: COLO 800, COLO 794, and A375 melanoma cell lines representing distinct metabolic phenotypes were analyzed. Targeted and untargeted metabolomics analyses integrated with Seahorse assays were performed to assess the effects of emodin and aloe-emodin on cell proliferation, mitochondrial function, and redox homeostasis. Glucose tracing using [U-13C6] glucose and metabolic flux analysis (MFA) were carried out to evaluate the glycolysis and TCA cycle dynamics. Results: Emodin and aloe-emodin inhibited proliferation by disrupting glycolysis, oxidative phosphorylation, and energy production across all cell lines. Both compounds impaired glucose metabolism, reduced TCA cycle intermediates, and induced mitochondrial ROS accumulation, causing oxidative stress and redox imbalance. Despite intrinsic metabolic differences, COLO 800 and COLO 794 upregulated antioxidant defenses; A375 enhanced one-carbon metabolism and amino acid pathways to maintain redox balance and nucleotide biosynthesis. Conclusions: Emodin and aloe-emodin can disrupt the metabolic plasticity of melanoma cells by impairing glycolysis, mitochondrial function, and redox homeostasis. Their ability to target metabolic vulnerabilities across diverse phenotypes highlights their therapeutic potential for overcoming resistance mechanisms and advancing melanoma treatment strategies. Full article
Show Figures

Figure 1

41 pages, 2908 KiB  
Review
Transcription Factor Blimp-1: A Central Regulator of Oxidative Stress and Metabolic Reprogramming in Chronic Inflammatory Diseases
by Aline Yen Ling Wang, Ana Elena Aviña, Yen-Yu Liu, Yun-Ching Chang and Huang-Kai Kao
Antioxidants 2025, 14(2), 183; https://doi.org/10.3390/antiox14020183 - 4 Feb 2025
Viewed by 1807
Abstract
B-lymphocyte-induced maturation protein 1 (Blimp-1) is a transcription factor that, among other functions, modulates metabolism and helps to regulate antioxidant pathways, which is important in the context of chronic inflammatory diseases like diabetes, cardiovascular disease, and autoimmune disease. In immune cell function, Blimp-1 [...] Read more.
B-lymphocyte-induced maturation protein 1 (Blimp-1) is a transcription factor that, among other functions, modulates metabolism and helps to regulate antioxidant pathways, which is important in the context of chronic inflammatory diseases like diabetes, cardiovascular disease, and autoimmune disease. In immune cell function, Blimp-1 has a modulatory role in the orchestration of metabolic reprogramming and as a promoter of anti-inflammatory cytokines, including IL-10, responsible for modulating oxidative stress and immune homeostasis. Moreover, Blimp-1 also modulates key metabolic aspects, such as glycolysis and fatty acid oxidation, which regulate reactive oxygen species levels, as well as tissue protection. This review depicts Blimp-1 as an important regulator of antioxidant defenses and anti-inflammation and suggests that the protein could serve as a therapeutic target in chronic inflammatory and metabolic dysregulation conditions. The modulation of Blimp-1 in diseases such as diabetic coronary heart disease and atherosclerosis could alleviate oxidative stress, augment the protection of tissues, and improve disease outcomes. The therapeutic potential for the development of new treatments for these chronic conditions lies in the synergy between the regulation of Blimp-1 and antioxidant therapies, which are future directions that may be pursued. This review emphasizes Blimp-1’s emerging importance as a novel regulator in the pathogenesis of inflammatory diseases, providing new opportunities for therapeutic intervention. Full article
(This article belongs to the Special Issue Oxidative Stress in Chronic Diseases: A Focus on Rheumatoid Arthritis)
Show Figures

Figure 1

25 pages, 2438 KiB  
Review
Radiotherapy Plus the Neurokinin-1 Receptor Antagonist Aprepitant: A Potent Therapeutic Strategy for the Treatment of Diffuse Intrinsic Pontine Glioma
by Miguel Muñoz and Marisa Rosso
Cancers 2025, 17(3), 520; https://doi.org/10.3390/cancers17030520 - 4 Feb 2025
Cited by 1 | Viewed by 1645
Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) is a devastating childhood brainstem tumor. The median survival of DIPG is 16–24 months independent of the treatment received. Therefore, new therapeutic strategies against DIPG are urgently needed. Substance P (SP) peptide, through the neurokinin neurokinin-1 [...] Read more.
Background: Diffuse intrinsic pontine glioma (DIPG) is a devastating childhood brainstem tumor. The median survival of DIPG is 16–24 months independent of the treatment received. Therefore, new therapeutic strategies against DIPG are urgently needed. Substance P (SP) peptide, through the neurokinin neurokinin-1 receptor (NK-1R), is involved in glioma progression. It induces glioma cell proliferation by activating MAPKs (p38 MAPK, ERK1/2, and JNK), c-Myc, AP-1, and NF-κB and induces antiapoptotic effects via PI3K/Akt/mTOR in glioma cells. SP favors glycogen breakdown that is essential for glycolysis. The SP/NK-1R system also regulates the migration and invasion of glioma cells, stimulates angiogenesis, and triggers inflammation which contributes to glioma progression. Moreover, all glioma cells express NK-1R, and NK-1R is essential for the viability of glioma cells and not of normal cells. In contrast, in glioma, NK-1R antagonists, such as the drug aprepitant, penetrate the brain and reach therapeutic concentrations, thereby inhibiting mitogenesis, inducing apoptosis, and inhibiting the breakdown of glycogen in glioma cells. In addition, they inhibit angiogenesis and exert antimetastatic and anti-inflammatory effects. The combination of radiotherapy with NK-1R antagonists produces radiosensitization and radioneuroprotection, reduces both peritumoral- and radiation-induced inflammation, and also provides antinausea and antivomiting effects. Objective: This review updates the involvement of the SP/NK-1R system in glioma promotion and progression and the potential clinical application of NK-1R antagonist drugs in DIPG therapy. Conclusions: NK-1R plays a crucial role in glioma progression and NK-1R antagonists such as aprepitant could be used in combination with radiotherapy as a potent therapeutic strategy for the treatment of patients with DIPG. Full article
(This article belongs to the Special Issue Outcomes in Glioblastoma Patients: From Diagnosis to Palliation)
Show Figures

Figure 1

18 pages, 713 KiB  
Review
Targeting Metabolic Vulnerabilities to Combat Drug Resistance in Cancer Therapy
by Taranatee Khan, Manojavan Nagarajan, Irene Kang, Chunjing Wu and Medhi Wangpaichitr
J. Pers. Med. 2025, 15(2), 50; https://doi.org/10.3390/jpm15020050 - 27 Jan 2025
Cited by 3 | Viewed by 2498
Abstract
Drug resistance remains a significant barrier to effective cancer therapy. Cancer cells evade treatment by reprogramming their metabolism, switching from glycolysis to oxidative phosphorylation (OXPHOS), and relying on alternative carbon sources such as glutamine. These adaptations not only enable tumor survival but also [...] Read more.
Drug resistance remains a significant barrier to effective cancer therapy. Cancer cells evade treatment by reprogramming their metabolism, switching from glycolysis to oxidative phosphorylation (OXPHOS), and relying on alternative carbon sources such as glutamine. These adaptations not only enable tumor survival but also contribute to immune evasion through mechanisms such as reactive oxygen species (ROS) generation and the upregulation of immune checkpoint molecules like PD-L1. This review explores the potential of targeting metabolic weaknesses in drug-resistant cancers to enhance therapeutic efficacy. Key metabolic pathways involved in resistance, including glycolysis, glutamine metabolism, and the kynurenine pathway, are discussed. The combination of metabolic inhibitors with immune checkpoint inhibitors (ICIs), particularly anti-PD-1/PD-L1 therapies, represents a promising approach to overcoming both metabolic and immune evasion mechanisms. Clinical trials combining metabolic and immune therapies have shown early promise, but further research is needed to optimize treatment combinations and identify biomarkers for patient selection. In conclusion, targeting cancer metabolism in combination with immune checkpoint blockade offers a novel approach to overcoming drug resistance, providing a potential pathway to improved outcomes in cancer therapy. Future directions include personalized treatments based on tumor metabolic profiles and expanding research to other tumor types. Full article
Show Figures

Graphical abstract

23 pages, 8721 KiB  
Review
The Physiological Functions and Therapeutic Potential of Hypoxia-Inducible Factor-1α in Vascular Calcification
by Zhenghong Zhang, Defan Wang, Renfeng Xu, Xiang Li, Zhengchao Wang and Yang Zhang
Biomolecules 2024, 14(12), 1592; https://doi.org/10.3390/biom14121592 - 12 Dec 2024
Cited by 1 | Viewed by 1892
Abstract
HIF-1α plays a crucial regulatory role in vascular calcification (VC), primarily influencing the osteogenic differentiation of VSMCs through oxygen-sensing mechanisms. Under hypoxic conditions, the stability of HIF-1α increases, avoiding PHD and VHL protein-mediated degradation, which promotes its accumulation in cells and then activates [...] Read more.
HIF-1α plays a crucial regulatory role in vascular calcification (VC), primarily influencing the osteogenic differentiation of VSMCs through oxygen-sensing mechanisms. Under hypoxic conditions, the stability of HIF-1α increases, avoiding PHD and VHL protein-mediated degradation, which promotes its accumulation in cells and then activates gene expressions related to calcification. Additionally, HIF-1α modulates the metabolic state of VSMCs by regulating the pathways that govern the switch between glycolysis and oxidative phosphorylation, thereby further advancing the calcification process. The interaction between HIF-1α and other signaling pathways, such as nuclear factor-κB, Notch, and Wnt/β-catenin, creates a complex regulatory network that serves as a critical driving force in VC. Therefore, a deeper understanding of the role and regulatory mechanism of the HIF-1α signaling during the development and progression of VC is of great significance, as it is not only a key molecular marker for understanding the pathological mechanisms of VC but also represents a promising target for future anti-calcification therapies. Full article
(This article belongs to the Special Issue Dysregulation of Calcium Signaling in Pathological Processes)
Show Figures

Figure 1

18 pages, 5556 KiB  
Article
Ascorbic Acid Enhances the Inhibitory Effect of Theasaponins against Candida albicans
by Yuhong Chen, Ying Gao and Junfeng Yin
Int. J. Mol. Sci. 2024, 25(19), 10661; https://doi.org/10.3390/ijms251910661 - 3 Oct 2024
Cited by 4 | Viewed by 1633
Abstract
Candida albicans (C. albicans) is a main cause of hospital-acquired fungal infections. Combination therapy is promising as a novel anti-C. albicans strategy because of its better efficacy. Theasaponins are pentacyclic triterpenes in the Camellia genus with multiple biological activities. Our [...] Read more.
Candida albicans (C. albicans) is a main cause of hospital-acquired fungal infections. Combination therapy is promising as a novel anti-C. albicans strategy because of its better efficacy. Theasaponins are pentacyclic triterpenes in the Camellia genus with multiple biological activities. Our previous studies prove that theasaponins display inhibitory activity against C. albicans. Ascorbic acid (VC) is a vitamin found in many plants that shows potential in combination therapy. However, whether VC enhances the activity of theasaponins remains unclear. In this study, the checkerboard micro-dilution method was used to assess the effect of VC (0–80 mmol/L) on the anti-C. albicans effect of theasaponins (0–1000 μg/mL). Then, the effects of theasaponins (31.25 μg/mL), VC (80 mmol/L), and theasaponins (31.25 μg/mL) + VC (80 mmol/L) on C. albicans planktonic cells and different stages of biofilm formation were assessed. Transcriptomic analysis was conducted to investigate the molecular mechanisms. According to the results, VC enhanced the anti-planktonic and anti-biofilm effect of theasaponins against C. albicans. The minimum inhibitory concentration of theasaponins was significantly decreased and the fungicidal efficiency was increased with the addition of VC. VC remarkably aggravated the suppression of theasaponins with regard to various virulence factors of C. albicans, including adhesion, early biofilm formation, mature biofilm, cell surface hydrophobicity, and phospholipase activity. Compared with the theasaponins or VC groups, the level of intracellular reactive oxygen species was higher, while the levels of mitochondrial membrane potential and adenosine triphosphate were lower in the combination group, suggesting more severe oxidative stress, mitochondrial injury, and energy deficiency. Transcriptomic analysis revealed that the combination predominantly suppressed the pathways of glycolysis, glycerophospholipid metabolism, glutathione metabolism, and cysteine and methionine metabolism. This implied that energy deficiency and redox imbalance were associated with the anti-C. albicans activity of the combination. These results prove that VC enhances the inhibitory effect of theasaponins against C. albicans and that the combination has the potential to be used as a topical antifungal therapy or disinfectant. Full article
(This article belongs to the Special Issue Antifungal Drug Discovery: Progresses, Challenges, Opportunities)
Show Figures

Figure 1

20 pages, 8986 KiB  
Article
Sanguinarine Induces Necroptosis of HCC by Targeting PKM2 Mediated Energy Metabolism
by Rui Kong, Nan Wang, Chunli Zhou, Yuqing Zhou, Xiaoyan Guo, Dongyan Wang, Yihai Shi, Rong Wan, Yuejuan Zheng and Jie Lu
Cancers 2024, 16(14), 2533; https://doi.org/10.3390/cancers16142533 - 13 Jul 2024
Cited by 2 | Viewed by 2135
Abstract
Backgrounds: Abnormal metabolism is the hallmark of hepatocellular carcinoma. Targeting energy metabolism has become the major focus of cancer therapy. The natural product, sanguinarine, displays remarkable anti-tumor properties by disturbing energy homeostasis; however, the underlying mechanism has not yet been elucidated. Methods: The [...] Read more.
Backgrounds: Abnormal metabolism is the hallmark of hepatocellular carcinoma. Targeting energy metabolism has become the major focus of cancer therapy. The natural product, sanguinarine, displays remarkable anti-tumor properties by disturbing energy homeostasis; however, the underlying mechanism has not yet been elucidated. Methods: The anticancer activity of sanguinarine was determined using CCK-8 and colony formation assay. Morphological changes of induced cell death were observed under electron microscopy. Necroptosis and apoptosis related markers were detected using western blotting. PKM2 was identified as the target by transcriptome sequencing. Molecular docking assay was used to evaluate the binding affinity of sanguinarine to the PKM2 molecule. Furthermore, Alb-CreERT2; PKM2loxp/loxp; Rosa26RFP mice was used to construct the model of HCC—through the intervention of sanguinarine in vitro and in vivo—to accurately explore the regulation effect of sanguinarine on cancer energy metabolism. Results: Sanguinarine inhibited tumor proliferation, metastasis and induced two modes of cell death. Molecular docking of sanguinarine with PKM2 showed appreciable binding affinity. PKM2 kinase activity and aerobic glycolysis rate declined, and mitochondrial oxidative phosphorylation was inhibited by sanguinarine application; these changes result in energy deficits and lead to necroptosis. Additionally, sanguinarine treatment prevents the translocation of PKM2 into the nucleus and suppresses the interaction of PKM2 with β-catenin; the transcriptional activity of PKM2/β-catenin signaling and its downstream genes were decreased. Conclusions: Sanguinarine showed remarkable anti-HCC activity via regulating energy metabolism by PKM2/β-catenin signaling. On the basis of these investigations, we propose that sanguinarine might be considered as a promising compound for discovery of anti-HCC drugs. Full article
Show Figures

Figure 1

16 pages, 3466 KiB  
Article
Restriction of Glycolysis Increases Serial Killing Capacity of Natural Killer Cells
by Lea Katharina Picard, Jens Alexander Niemann, Elisabeth Littwitz-Salomon, Herbert Waldmann and Carsten Watzl
Int. J. Mol. Sci. 2024, 25(5), 2917; https://doi.org/10.3390/ijms25052917 - 2 Mar 2024
Cited by 3 | Viewed by 2677
Abstract
Tumor cells rely heavily on glycolysis to meet their high metabolic demands. While this results in nutrient deprivation within the tumor microenvironment and has negative effects on infiltrating immune cells such as natural killer (NK) cells, it also creates a potential target for [...] Read more.
Tumor cells rely heavily on glycolysis to meet their high metabolic demands. While this results in nutrient deprivation within the tumor microenvironment and has negative effects on infiltrating immune cells such as natural killer (NK) cells, it also creates a potential target for cancer therapies. Here we use Glupin, an inhibitor of glucose transporters, to study the effect of limited glucose uptake on NK cells and their anti-tumor functions. Glupin treatment effectively inhibited glucose uptake and restricted glycolysis in NK cells. However, acute treatment had no negative effect on NK cell cytotoxicity or cytokine production. Long-term restriction of glucose uptake via Glupin treatment only delayed NK cell proliferation, as they could switch to glutaminolysis as an alternative energy source. While IFN-γ production was partially impaired, long-term Glupin treatment had no negative effect on degranulation. Interestingly, the serial killing activity of NK cells was even slightly enhanced, possibly due to changes in NAD metabolism. This demonstrates that NK cell cytotoxicity is remarkably robust and insensitive to metabolic disturbances, which makes cellular metabolism an attractive target for immune-mediated tumor therapies. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

15 pages, 13944 KiB  
Article
Atractylenolide II Suppresses Glycolysis and Induces Apoptosis by Blocking the PADI3-ERK Signaling Pathway in Endometrial Cancer Cells
by Shuang Tian, Lili Ren, Chao Liu and Zhe Wang
Molecules 2024, 29(5), 939; https://doi.org/10.3390/molecules29050939 - 21 Feb 2024
Cited by 5 | Viewed by 2165
Abstract
Atractylenolide II (AT-II), the major bioactive compound of Atractylodes macrocephala, exhibits anti-cancer activity against many types of tumors, but the roles and the potential mechanisms in endometrial cancer remain unclear. In the present study, AT-II treatment was found to significantly suppress RL95-2 [...] Read more.
Atractylenolide II (AT-II), the major bioactive compound of Atractylodes macrocephala, exhibits anti-cancer activity against many types of tumors, but the roles and the potential mechanisms in endometrial cancer remain unclear. In the present study, AT-II treatment was found to significantly suppress RL95-2 and AN3CA cell proliferation and glycolysis, and induced their apoptosis by inactivating the ERK signaling pathway, accompanied by the changing expression of the glycolytic key enzymes and apoptotic-related proteins. Peptidyl arginine deiminase 3 (PADI3), as the candidate target gene of AT-II, was highly expressed in the endometrial cancer tissues and associated with a poor prognosis according to bioinformatics analysis. PADI3 knockdown inhibited proliferation and glycolysis in endometrial cancer cells and induced cell apoptosis. Furthermore, AT-II negatively regulated the expression of PADI3, and PADI3 overexpression reversed the effects of AT-II on endometrial cancer cells. Our findings suggested that the anti-cancer function of AT-II is associated with the suppression of glycolysis and induction of apoptosis by blocking the PADI3-ERK signaling pathway. Thus, AT-II represents a novel therapeutic target for endometrial cancer and targeting AT-II may serve as a potential strategy for the clinical therapy of endometrial cancer. Full article
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)
Show Figures

Figure 1

Back to TopTop