51
|
Chen X, Kadier M, Shi M, Li K, Chen H, Xia Y, Wang Q, Li R, Long Y, Qin J, Wang H, Jiang G. Targeting Melatonin to Mitochondria Mitigates Castration-Resistant Prostate Cancer by Inducing Pyroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2408996. [PMID: 40285589 DOI: 10.1002/smll.202408996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Prostate cancer frequently progresses to castration-resistant prostate cancer (CRPC) following androgen deprivation therapy, presenting a significant clinical challenge. Targeting tumor metabolism, particularly mitochondrial pathways, offers a promising strategy for overcoming CRPC. The modification of melatonin (Mel) to a triphenylphosphonium (TPP) cation-targeted mitochondria-melatonin (Mito-Mel) significantly increases its potency by over 1000-fold. Mito-Mel selectively targets mitochondria, enhancing reactive oxygen species (ROS) generation and causing mitochondrial membrane potential disruption. This leads to the inhibition of mitochondrial respiration including the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS), which, in turn, suppresses CRPC survival metabolic adaptations, such as glycolysis. In vitro and in vivo experiments reveal for the first time that natural small molecule compound with mitochondrial targeting via TPP exhibits excellent anticancer efficacy by inducing tumor cellular pyroptosis and facilitating the immune response, underlining the encouraging promise of this strategy for the effective treatment of CRPC.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Mairehaba Kadier
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Mengting Shi
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Kefeng Li
- Center for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macau SAR, 999078, China
| | - Hongtao Chen
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Yongzhen Xia
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Qiaohua Wang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Rongna Li
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Yili Long
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Jingbo Qin
- Guangdong Provincial Engineering Research Center of Molecular Imaging and Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine Foundation of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Hao Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| |
Collapse
|
52
|
Cui J, Liu W, Zhong S, Fang Y, Xu P, Xu C, Wang R, Hu X, Zhou W, Li K, Hong M, Qian S, Sun Q. Blockade of TIGAR prevents CD8 + T cell dysfunction and elicits anti-AML immunity. Cancer Immunol Immunother 2025; 74:183. [PMID: 40285889 PMCID: PMC12033161 DOI: 10.1007/s00262-025-04042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Acute myeloid leukemia (AML) cells and activated T cells rely on aerobic glycolysis for energy metabolism. The TP53-induced glycolysis and apoptosis regulator (TIGAR) inhibits glycolysis and protects AML cells from apoptosis. Preliminary studies suggest that combining TIGAR inhibition with the glycolysis inhibitor 2-deoxy-D-glucose (2-DG) may offer a therapeutic strategy for AML. However, it remains unclear whether silencing TIGAR can enhance T cell function and thereby improve AML prognosis. This study aims to investigate whether TIGAR silencing in host can eliminate AML cells and rejuvenate dysfunctional T cells with mouse models. TIGAR knockout mice on the C57BL/6J background were generated and AML mouse models were established through intravenous injection of C1498 cells. We found that TIGAR depletion enhanced CD8+ T cell counts and raised CD4/CD8 ratio, downregulating CD44 and immune checkpoints CTLA-4, LAG-3, PD-1 on cell surface of CD8+ T cells. TIGAR depletion boosted cytokine secretion (IFN-γ, perforin, granzyme B, TNF-α) by CD8+ T cells and IL-2, TNF-α by CD4+ T cells, improving cytotoxicity against AML cells, proliferation, and reducing apoptosis. TIGAR suppression in host with 2-DG prolonged AML mouse survival, decreased tumor burden, and leukemic infiltration. TIGAR suppression restored thymic T cell development and peripheral immune balance. Single-cell RNA sequencing analysis also revealed that high TIGAR expression influences the glycolysis pathway, and correlates with markers of T cell exhaustion. This study indicates that blocking TIGAR prevents CD8+ T cell dysfunction and induces anti-AML immunity.
Collapse
Affiliation(s)
- Jialin Cui
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wenjie Liu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Shiyang Zhong
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, China
| | - Yiran Fang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Pei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Cheng Xu
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Rong Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Xingfei Hu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, China
| | - Wanting Zhou
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, China
| | - Ming Hong
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Sixuan Qian
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| | - Qian Sun
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
53
|
Wei J, Mo CD, Zhu GY, Qiu FY, Qin SP, Hou QY, Zhou Y, Wei WS, Huang Z, Yang JR. The relationship between infectious pathogen antibodies, plasma metabolites, and breast cancer: A Mendelian randomization study with mediation analysis. Medicine (Baltimore) 2025; 104:e42283. [PMID: 40295253 PMCID: PMC12040016 DOI: 10.1097/md.0000000000042283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) has the second highest incidence rate among women worldwide. Although there are various treatment methods, the prognosis is poor once metastasis occurs. However, the extent to which pathogens of infectious diseases influence the risk of BC remains unclear. The goal of this study is to determine if these pathogens are causally related to BC development. A Mendelian randomization (MR) analysis is used to assess the causal relationship between infectious pathogen antibodies and the risk of BC, as well as their potential intermediary factors. Two-sample MR analysis using inverse variance weighting (IVW) is conducted to determine the causal relationship between infectious pathogen antibodies and the risk of BC. Additionally, the mediating role of 1400 metabolites between infectious pathogen antibodies and the risk of BC is analyzed. There were 5 infectious pathogen antibodies and 86 metabolites associated with BC (P < .05). There were 14 metabolites that mediated the pathway between infectious pathogen antibodies and BC. X-07765 levels showed a significant negative mediating effect on the relationship between Anti-human herpes virus 6 IgG seropositivity and BC (beta = -0.0025, 95% CI [-0.0046, -0.0003], P = .0236), accounting for 14.8% of the effect (95% CI: 27.7-1.99). This study provides a thorough evaluation of the causal relationships among infectious pathogen antibodies, plasma metabolites, and BC. Our research has identified 5 infectious pathogen antibodies that exhibit a causal relationship with BC, mediated through 86 distinct metabolites.
Collapse
Affiliation(s)
- Jing Wei
- Guangxi Medical University, Nanning, PR China
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Chong-de Mo
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Gui-yue Zhu
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Fang-yu Qiu
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Sheng-peng Qin
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Qi-yan Hou
- Graduate School of Guangxi University of Chinese Medicine,Nanning, PR China
| | - Ying Zhou
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Wen-song Wei
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Zhen Huang
- Department of Breast Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| | - Jian-rong Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, PR China
| |
Collapse
|
54
|
Fukunaga H, Fukunaga M. Mitochondrial DNA copy numbers in gastric cancer tissues: a possible biomarker for estimating cancer progression. Jpn J Clin Oncol 2025:hyaf066. [PMID: 40263745 DOI: 10.1093/jjco/hyaf066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Mitochondria have their own genome (mtDNA), which in humans is a circular multi-copy genome consisting of 16 569 base pairs. Abnormalities in the mtDNA have been reported to correlate with various age-related pathophysiologies. METHODS Based on a total of 182 DNA samples extracted from gastric cancer tissues, we measured mtDNA copy numbers (mtDNA-CN) using real-time polymerase chain reaction (PCR) and then examined alongside sex, age, tumor stage, Laurén classification, and the overexpression of Human Epidermal Growth Factor Receptor 2 (HER2). RESULTS We found no sex differences in mtDNA-CN and no correlation with age, but significant differences according to tumor stage. The mtDNAcn of intestinal type by Laurén classification was significantly larger than that of diffuse type. There was no significant difference in mtDNA-CN between HER2-positive and -negative tissues. Multiple regression analyses showed that only the tumor stage was a significant variable, while Laurén classification was not. CONCLUSION These results indicate that mitochondrial genomic abnormalities contribute the progression of gastric cancer independently of HER2 overexpression, and may shed light on the emerging role of mtDNA-CN in situ as a possible biomarker for estimating cancer progression.
Collapse
Affiliation(s)
- Hisanori Fukunaga
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, N12 W5 Kita-ku, Sapporo 060-0812, Japan
| | - Mayuko Fukunaga
- Department of Internal Medicine, Sapporo Daiichi Hospital, Niju-Yonken 4-jo 3-chome 4-26, Nishi-ku, Sapporo 063-0804, Japan
- Department of Gastroenterology and Hepatology, School of Medicine, Sapporo Medical University, S1 W16 Chuo-ku, Sapporo 060-8543, Japan
| |
Collapse
|
55
|
Cao L, Leclercq-Cohen G, Klein C, Sorrentino A, Bacac M. Mechanistic insights into resistance mechanisms to T cell engagers. Front Immunol 2025; 16:1583044. [PMID: 40330489 PMCID: PMC12053166 DOI: 10.3389/fimmu.2025.1583044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
T cell engagers (TCEs) represent a groundbreaking advancement in the treatment of B and plasma cell malignancies and are emerging as a promising therapeutic approach for the treatment of solid tumors. These molecules harness T cells to bind to and eliminate cancer cells, effectively bypassing the need for antigen-specific T cell recognition. Despite their established clinical efficacy, a subset of patients is either refractory to TCE treatment (e.g. primary resistance) or develops resistance during the course of TCE therapy (e.g. acquired or treatment-induced resistance). In this review we comprehensively describe the resistance mechanisms to TCEs, occurring in both preclinical models and clinical trials with a particular emphasis on cellular and molecular pathways underlying the resistance process. We classify these mechanisms into tumor intrinsic and tumor extrinsic ones. Tumor intrinsic mechanisms encompass changes within tumor cells that impact the T cell-mediated cytotoxicity, including tumor antigen loss, the expression of immune checkpoint inhibitory ligands and intracellular pathways that render tumor cells resistant to killing. Tumor extrinsic mechanisms involve factors external to tumor cells, including the presence of an immunosuppressive tumor microenvironment (TME) and reduced T cell functionality. We further propose actionable strategies to overcome resistance offering potential avenues for enhancing TCE efficacy in the clinic.
Collapse
Affiliation(s)
- Linlin Cao
- Roche Innovation Center, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
56
|
Wang T, Brown C, Doherty N, Byrne NM, Islam R, Doherty M, Feng J, Yin C, Chambers S, McQuoid L, Mohamed-Smith L, Butterworth KT, Kerr EM, Coulter JA. Mannose and PMI depletion overcomes radiation resistance in HPV-negative head and neck cancer. Cell Commun Signal 2025; 23:189. [PMID: 40259370 PMCID: PMC12013184 DOI: 10.1186/s12964-025-02204-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025] Open
Abstract
Radiotherapy is critical component of multidisciplinary cancer care, used as a primary and adjuvant treatment for patients with head and neck squamous cell carcinoma. This study investigates how mannose, a naturally occurring monosaccharide, combined with phosphomannose isomerase (PMI) depletion, enhances the sensitivity of HPV-negative head and neck tumour models to radiation. Isogenic PMI knockout models were generated by CRISPR/Cas9 gene editing, yielding a 20-fold increase in sensitivity to mannose in vitro, and causing significant tumour growth delay in vivo. This effect is driven by metabolic reprogramming, resulting in potent glycolytic suppression coupled with consistent depletion of ATP and glycolytic intermediates in PMI-depleted models. Functionally, these changes impede DNA damage repair following radiation, resulting in a significant increase in radiation sensitivity. Mannose and PMI ablation supressed both oxygen consumption rate and extracellular acidification, pushing cells towards a state of metabolic quiescence, effects contributing to increased radiation sensitivity under both normoxic and hypoxic conditions. In 3D-tumoursphere models, metabolic suppression by mannose and PMI depletion was shown to elevate intra-tumoursphere oxygen levels, contributing to significant in vitro oxygen-mediated radiosensitisation. These findings position PMI as a promising anti-tumour target, highlighting the potential of mannose as a metabolic radiosensitiser enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
- Tongchuan Wang
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Connor Brown
- Patrick G. Johnston Centre for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Niamh Doherty
- Patrick G. Johnston Centre for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Niall M Byrne
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Rayhanul Islam
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Meabh Doherty
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Jie Feng
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Cancan Yin
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Sarah Chambers
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Lydia McQuoid
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Letitia Mohamed-Smith
- Patrick G. Johnston Centre for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Karl T Butterworth
- Patrick G. Johnston Centre for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Emma M Kerr
- Patrick G. Johnston Centre for Cancer Research, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Jonathan A Coulter
- School of Pharmacy, Queens University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
57
|
Addanki S, Kim L, Stevens A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers (Basel) 2025; 17:1355. [PMID: 40282531 PMCID: PMC12025543 DOI: 10.3390/cancers17081355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by aggressive proliferation and metabolic reprogramming that support its survival and resistance to therapy. This review explores the metabolic distinctions between AML cells and normal hematopoietic stem cells (HSCs), emphasizing the role of altered mitochondrial function, oxidative phosphorylation (OXPHOS), and biosynthetic pathways in leukemic progression. AML cells exhibit distinct metabolic vulnerabilities, including increased mitochondrial biogenesis, reliance on glycolysis and amino acid metabolism, and unique signaling interactions that sustain leukemic stem cells (LSCs). These dependencies provide potential therapeutic targets, as metabolic inhibitors have demonstrated efficacy in disrupting AML cell survival while sparing normal hematopoietic cells. We examine current and emerging metabolic therapies, such as inhibitors targeting glycolysis, amino acid metabolism, and lipid biosynthesis, highlighting their potential in overcoming drug resistance. However, challenges remain in translating these strategies into clinical practice due to AML's heterogeneity and adaptability. Further research into AML's metabolic plasticity and precision medicine approaches is crucial for improving treatment outcomes. Understanding and exploiting AML's metabolic vulnerabilities could pave the way for novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Sridevi Addanki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Alexandra Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
58
|
Gamal W, Mediavilla-Varela M, Kunta V, Sahakian E, Pinilla-Ibarz J. Impact of mitochondrial metabolism on T-cell dysfunction in chronic lymphocytic leukemia. Front Cell Dev Biol 2025; 13:1577081. [PMID: 40313718 PMCID: PMC12043688 DOI: 10.3389/fcell.2025.1577081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
T cells play a central role in anti-tumor immunity, yet their function is often compromised within the immunosuppressive tumor microenvironment, leading to cancer progression and resistance to immunotherapies. T-cell activation and differentiation require dynamic metabolic shifts, with mitochondrial metabolism playing a crucial role in sustaining their function. Research in cancer immunometabolism has revealed key mitochondrial abnormalities in tumor-infiltrating lymphocytes, including reduced mitochondrial capacity, depolarization, structural defects, and elevated reactive oxygen species. While these mitochondrial disruptions are well-characterized in solid tumors and linked to T-cell exhaustion, their impact on T-cell immunity in lymphoproliferative disorders remains underexplored. Chronic lymphocytic leukemia (CLL), the most prevalent chronic adult leukemia, is marked by profound T-cell dysfunction that limits the success of adoptive cell therapies. Emerging studies are shedding light on the role of mitochondrial disturbances in CLL-related T-cell dysfunction, but significant knowledge gaps remain. This review explores mitochondrial metabolism in T-cell exhaustion, emphasizing recent findings in CLL. We also discuss therapeutic strategies to restore T-cell mitochondrial function and identify key research gaps.
Collapse
Affiliation(s)
- Wael Gamal
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Melanie Mediavilla-Varela
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Vishaal Kunta
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
59
|
Ghanbarian M, Dolgova N, Vizeacoumar FS, Vizeacoumar FJ, Michel D, El-Aneed A, Dmitriev OY. Metabolic Effects of the Cancer Metastasis Modulator MEMO1. Metabolites 2025; 15:277. [PMID: 40278406 PMCID: PMC12029338 DOI: 10.3390/metabo15040277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Cancer cells often display altered energy metabolism. In particular, expression levels and activity of the tricarboxylic acid cycle (TCA cycle) enzymes may change in cancer, and dysregulation of the TCA cycle is a frequent hallmark of cancer cell metabolism. MEMO1, a modulator of cancer metastasis, has been shown to bind iron and regulate iron homeostasis in the cells. MEMO1 knockout changed mitochondrial morphology and iron content in breast cancer cells. Our previous genome-wide analysis of MEMO1 genetic interactions across multiple cancer cell lines revealed that gene sets involved in mitochondrial respiration and the TCA cycle are enriched among the gain-of-function interaction partners of MEMO1. Based on these findings, we measured the TCA cycle metabolite levels in breast cancer cells with varying levels of MEMO1 expression. Methods: ShRNA knockdown assay was performed to test essentiality of key TCA cycle enzymes. TCA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in MDA-MB-231 (high MEMO1), M67-2 (MEMO1 knockdown), and M67-9 (MEMO1 knockout) cells under iron-depleted, basal iron, and iron-supplemented conditions. Results:ACO2 and OGDH knockdowns inhibit cell proliferation, indicating an essential role of the TCA cycle in MDA-MB-231 metabolism. α-Ketoglutarate and citrate levels exhibited an inverse relationship with MEMO1 expression, increasing significantly in MEMO1 knockout cells regardless of iron availability. In contrast, fumarate, malate, and glutamate levels were elevated in MEMO1 knockout cells specifically under low iron conditions, suggesting an iron-dependent effect. Conclusions: Overall, our results indicate that MEMO1 plays a role in regulating the TCA in cancer cells in an iron-dependent manner.
Collapse
Affiliation(s)
- Marziyeh Ghanbarian
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| | - Natalia Dolgova
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| | - Frederick S. Vizeacoumar
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Franco J. Vizeacoumar
- Cancer Research Department, Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
- Division of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Deborah Michel
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anas El-Aneed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Oleg Y. Dmitriev
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| |
Collapse
|
60
|
Chen Y, Zhu H, Luo Y, Xie T, Hu Y, Yan Z, Ji W, Wang Y, Yin Q, Xian H. ALDOC promotes neuroblastoma progression and modulates sensitivity to chemotherapy drugs by enhancing aerobic glycolysis. Front Immunol 2025; 16:1573815. [PMID: 40313939 PMCID: PMC12043483 DOI: 10.3389/fimmu.2025.1573815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Neuroblastoma (NB), a malignant extracranial solid tumor originating from the sympathetic nervous system, exhibits poor prognosis in high-risk cases, with a 5-year overall survival rate below 50%. Glycolysis has been implicated in NB pathogenesis, and targeting glycolysis-related pathways shows therapeutic potential. This study investigates the role of the glycolysis-associated gene ALDOC in NB pathogenesis and its impact on chemotherapy sensitivity. Methods Transcriptomic data from NB patients were analyzed to identify ALDOC as an independent risk factor for high-risk NB. Protein expression levels of ALDOC were assessed in NB cells versus normal cells using immunoblotting. Functional experiments, including proliferation and migration assays, were conducted in ALDOC-interfered NB cell lines. Glycolytic activity was evaluated by measuring glucose uptake, lactate production, and ATP generation. Additionally, the sensitivity of ALDOC-downregulated NB cells to cisplatin and cyclophosphamide was tested to explore its role in chemotherapy response. Results ALDOC was identified as a high-risk prognostic marker in NB, with elevated protein expression in NB cells compared to normal controls. Silencing ALDOC significantly inhibited NB cell proliferation and migration. Glycolytic activity was markedly reduced in ALDOC-downregulated cells, evidenced by decreased glucose uptake, lactate production, and ATP levels. Furthermore, ALDOC suppression enhanced NB cell sensitivity to cisplatin and cyclophosphamide, suggesting a glycolysis-dependent mechanism underlying chemotherapy resistance. Discussion Our findings highlight ALDOC as a critical driver of NB progression through glycolysis acceleration, with implications for therapeutic targeting. The observed increase in chemotherapy sensitivity upon ALDOC inhibition underscores its potential as a biomarker for treatment optimization. However, the complexity of glycolysis regulation, involving multiple genes and pathways, necessitates further mechanistic studies to clarify ALDOC's specific role. Despite this limitation, our work emphasizes the importance of aerobic glycolysis in NB pathogenesis and provides a foundation for developing novel therapeutic strategies targeting ALDOC or associated pathways. Future research should explore interactions between ALDOC and other glycolytic regulators to refine combinatorial treatment approaches.
Collapse
Affiliation(s)
- Yunpeng Chen
- School of Medicine, Nantong University, Nantong, China
| | - Haixia Zhu
- Cancer Research Center Nantong, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Yishu Luo
- School of Medicine, Nantong University, Nantong, China
| | - Tianyue Xie
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, China
| | - Youyang Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhiwei Yan
- School of Medicine, Nantong University, Nantong, China
| | - Weichao Ji
- School of Medicine, Nantong University, Nantong, China
| | - YaXuan Wang
- Department of Urology, Nantong Tumor Hospital, Nantong, China
| | - Qiyou Yin
- Department of Paediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hua Xian
- Department of Paediatric Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
61
|
Hayashi M, Nakamura K, Harada S, Tanaka M, Kobayashi A, Saito H, Tsuji T, Yamamoto D, Moriyama H, Kinoshita J, Inaki N. GLUT1 inhibition by BAY-876 induces metabolic changes and cell death in human colorectal cancer cells. BMC Cancer 2025; 25:716. [PMID: 40247224 PMCID: PMC12004878 DOI: 10.1186/s12885-025-14141-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Glucose transporter 1 (GLUT1) is known to play a crucial role in glucose uptake in malignant tumors. GLUT1 inhibitors reportedly exhibit anti-tumor effects by suppressing cancer cell proliferation. BAY-876, a selective GLUT1 inhibitor, has been shown to inhibit tumor growth in ovarian and breast cancers. In this study, we investigated the anti-proliferative effects of BAY-876 treatment in human colorectal cancer (CRC) cell lines. METHODS We investigated the metabolic changes and effects on proliferation from BAY-876 treatment in HCT116, DLD1, COLO205, LoVo, and Caco-2 cells in vitro. Additionally, a mouse xenograft model was established using HCT116 cells to examine the tumor-inhibitory effects of BAY-876 treatment in vivo. RESULTS BAY-876 treatment inhibited cell proliferation in HCT116, DLD1, COLO205, and LoVo cells. Reduced GLUT1 protein expression levels were observed through western blot analysis. Flux analysis indicated enhanced mitochondrial respiration, accompanied by increased reactive oxygen species levels and apoptosis rates. Tumor-inhibitory effects were also observed in the xenograft model, with the BAY-876-treated groups showing GLUT1 suppression. CONCLUSIONS BAY-876 treatment induced metabolic changes and inhibited cell proliferation in human CRC cell lines. Using BAY-876 is a potential novel approach for treating CRC.
Collapse
Affiliation(s)
- Masato Hayashi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Keishi Nakamura
- Department of Surgery, Public Central Hospital of Matto Ishikawa, 3-8 Kuramitsu, Hakusan, Ishikawa, 924-8588, Japan.
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Mariko Tanaka
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Akiko Kobayashi
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroto Saito
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Toshikatsu Tsuji
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Daisuke Yamamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hideki Moriyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Noriyuki Inaki
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
62
|
Li H, Jia F, Wang X, Yang T, Wang JH. Efficient and Discriminative Isolation of Circulating Cancer Stem Cells and Non-Stem-like Circulating Tumor Cells Using a Click-Handle-Loaded M13 Phage-Based Surface. Anal Chem 2025; 97:8080-8087. [PMID: 40192481 DOI: 10.1021/acs.analchem.5c00924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) are crucial for cancer research and clinical applications, with circulating cancer stem cells (cCSCs) being a rare but key subpopulation responsible for metastasis, recurrence, and therapy resistance. Current limitations in efficiently isolating these cells, particularly distinguishing cCSCs from non-stem-like CTCs (nsCTCs), hinder our understanding of cancer progression and precision medicine strategies. Herein, we developed a novel CTC isolation approach that integrates cell metabolic chemical tagging with a click-handle-loaded M13 phage-based surface (CHPhace). The multivalent nature of flexible M13 nanofibers, featuring thousands of modification sites for click reactions, significantly enhances CTC capture across diverse tumor types. Leveraging the unique slow-cycling characteristic of cCSCs, CHPhace demonstrated selective cCSCs isolation through metabolic labeling and demetabolism processes. The robust performance of CHPhace allows efficient isolation of both cCSCs and nsCTCs from complex blood sample matrices, achieving capture efficiencies exceeding 80%. This approach represents a promising tool for advancing our understanding of cancer progression and enhancing precision in clinical diagnosis and cancer prognosis.
Collapse
Affiliation(s)
- Huida Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Fengting Jia
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xin Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
63
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
64
|
Abdeen SK, Mastandrea I, Stinchcombe N, Puschhof J, Elinav E. Diet-microbiome interactions in cancer. Cancer Cell 2025; 43:680-707. [PMID: 40185096 DOI: 10.1016/j.ccell.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
Diet impacts cancer in diverse manners. Multiple nutritional effects on tumors are mediated by dietary modulation of commensals, residing in mucosal surfaces and possibly also within the tumor microenvironment. Mechanistically understanding such diet-microbiome-host interactions may enable to develop precision nutritional interventions impacting cancer development, dissemination, and treatment responses. However, data-driven nutritional strategies integrating diet-microbiome interactions are infrequently incorporated into cancer prevention and treatment schemes. Herein, we discuss how dietary composition affects cancer-related processes through alterations exerted by specific nutrients and complex foods on the microbiome. We highlight how dietary timing, including time-restricted feeding, impacts microbial function in modulating cancer and its therapy. We review existing and experimental nutritional approaches aimed at enhancing microbiome-mediated cancer treatment responsiveness while minimizing adverse effects, and address challenges and prospects in integrating diet-microbiome interactions into precision oncology. Collectively, mechanistically understanding diet-microbiome-host interactomes may enable to achieve a personalized and microbiome-informed optimization of nutritional cancer interventions.
Collapse
Affiliation(s)
- Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nina Stinchcombe
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Junior Research Group Epithelium Microbiome Interactions, DKFZ, Heidelberg, Germany
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Junior Research Group Epithelium Microbiome Interactions, DKFZ, Heidelberg, Germany.
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
65
|
Caravia LG, Mitranovici MI, Oala IE, Tiron AT, Simionescu AA, Borcan AM, Craina M. The Importance of Cancer Stem Cells and Their Pathways in Endometrial Cancer: A Narrative Review. Cells 2025; 14:594. [PMID: 40277919 PMCID: PMC12025850 DOI: 10.3390/cells14080594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Endometrial cancer is one of the most common malignancies seen in women in developed countries. While patients in the early stages of this cancer show better responses to surgery, adjuvant hormonal therapy, and chemotherapy, patients with recurrence show treatment resistance. Researchers have recently focused on cancer stem cells (CSCs) in the treatment of gynecologic cancer in general but also specifically in endometrial cancer. CSCs have been investigated because of their resistance to conventional therapies, such as chemo- and radiotherapy, and their ability to induce the progression and recurrence of malignancy. The activation of alternative pathways, such as WNT, PI3K, NF-kB, or NOTCH, could be the basis of the acquisition of these abilities of CSCs. Their specific markers and signaling pathways could be treatment targets for CSCs. In this article, we discuss the importance of obtaining a better understanding of the molecular basis and pathways of CSCs in endometrial cancer and the role of CSCs, aiming to discover more specific therapeutic approaches.
Collapse
Affiliation(s)
- Laura Georgiana Caravia
- Division of Cellular and Molecular Biology and Histology, Department of Morphological Sciences, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Melinda Ildiko Mitranovici
- Public Health Department, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Ioan Emilian Oala
- Department of Obstetrics and Gynecology, Emergency County Hospital Hunedoara, 14 Victoriei Street, 331057 Hunedoara, Romania;
| | - Andreea Taisia Tiron
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Anca Angela Simionescu
- Department of Obstretics and Gynecology, Filantropia, Faculty of Medicine Carol Davila, 011171 Bucharest, Romania;
| | - Alina Maria Borcan
- Department of Microbiology, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, Faculty of Medicine Carol Davila, 021105 Bucharest, Romania;
| | - Marius Craina
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
66
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
67
|
Yan W, Saqirile, Li K, Li K, Wang C. The Role of N6-Methyladenosine in Mitochondrial Dysfunction and Pathology. Int J Mol Sci 2025; 26:3624. [PMID: 40332101 PMCID: PMC12026702 DOI: 10.3390/ijms26083624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Mitochondria are indispensable in cells and play crucial roles in maintaining cellular homeostasis, energy production, and regulating cell death. Mitochondrial dysfunction has various manifestations, causing different diseases by affecting the diverse functions of mitochondria in the body. Previous studies have mainly focused on mitochondrial-related diseases caused by nuclear gene mutations or mitochondrial gene mutations, or mitochondrial dysfunction resulting from epigenetic regulation, such as DNA and histone modification. In recent years, as a popular research area, m6A has been involved in a variety of important processes under physiological and pathological conditions. However, there are few summaries on how RNA methylation, especially m6A RNA methylation, affects mitochondrial function. Additionally, the role of m6A in pathology through influencing mitochondrial function may provide us with a new perspective on disease treatment. In this review, we summarize several manifestations of mitochondrial dysfunction and compile examples from recent years of how m6A affects mitochondrial function and its role in some diseases.
Collapse
Affiliation(s)
| | | | | | | | - Changshan Wang
- School of Life Science, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (S.); (K.L.); (K.L.)
| |
Collapse
|
68
|
Zehetner L, Széliová D, Kraus B, Hernandez Bort JA, Zanghellini J. Multi-omics driven genome-scale metabolic modeling improves viral vector yield in HEK293. Metab Eng 2025; 91:103-118. [PMID: 40220853 DOI: 10.1016/j.ymben.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/06/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
HEK293 cells are a versatile cell line extensively used in the production of recombinant proteins and viral vectors, notably Adeno-associated virus (AAV) (Bulcha et al., 2021). Despite their high transfection efficiency and adaptability to various culture conditions, challenges remain in achieving sufficient yields of active viral particles. This study presents a comprehensive multi-omics analysis of two HEK293 strains under good manufacturing practice conditions, focusing on the metabolic and cellular responses during AAV production. The investigation included lipidomic, exometabolomic, and transcriptomic profiling across different conditions and time points. Genome-scale metabolic models (GSMMs) were reconstructed for these strains to elucidate metabolic shifts and identify potential bottlenecks in AAV production. Notably, the study revealed significant differences between a High-producing (HP) and a Low-producing (LP) HEK293 strains, highlighting pseudohypoxia in the LP strain. Key findings include the identification of hypoxia-inducible factor 1-alpha (HIF-1α) as a critical regulator in the LP strain, linking pseudohypoxia to poor AAV productivity. Inhibition of HIF-1α resulted in immediate cessation of cell growth and a 2.5-fold increase in viral capsid production, albeit with a decreased number of viral genomes, impacting the full-to-empty particle ratio. This trade-off is significant because it highlights a key challenge in AAV production: achieving a balance between capsid assembly and genome packaging to optimize the yield of functional viral vectors. Overall this suggests that while HIF-1α inhibition enhances capsid assembly, it simultaneously hampers nucleotide synthesis via the pentose phosphate pathway (PPP), necessary for nucleotide synthesis, and therefore for AAV genome replication.
Collapse
Affiliation(s)
- L Zehetner
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria; Doctoral School of Chemistry, University of Vienna, Vienna, 1090, Austria.
| | - D Széliová
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria.
| | - B Kraus
- Institute of Molecular Biotechnology, Institut für Molekulare Biotechnologie GmbH, Vienna, 1030, Austria
| | - J A Hernandez Bort
- Department of Applied Life Sciences, Bioengineering, University of Applied Sciences Campus Vienna, Vienna, 1100, Austria.
| | - J Zanghellini
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
69
|
Salnikov OG, Chukanov NV, Pravdivtsev AN, Burueva DB, Sviyazov SV, Them K, Hövener JB, Koptyug IV. Heteronuclear Parahydrogen-Induced Hyperpolarization via Side Arm Hydrogenation. Chemphyschem 2025:e2401119. [PMID: 40211662 DOI: 10.1002/cphc.202401119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/22/2025] [Indexed: 05/02/2025]
Abstract
Nuclear spin hyperpolarization dramatically enhances the sensitivity of nuclear magnetic resonance spectroscopy and imaging. Hyperpolarization of biomolecules (e.g., pyruvate) is of particular interest as it allows one to follow their metabolism, providing a diagnostic tool for various pathologies, including cancer. In this regard, the hyperpolarization of 13C nuclei is especially beneficial due to its typically relatively long hyperpolarization lifetime and the absence of a background signal. Parahydrogen-induced polarization (PHIP) is arguably the most affordable hyperpolarization technique. PHIP exploits the pairwise addition of parahydrogen to an unsaturated substrate. This sets limitations on the range of compounds amenable to direct PHIP hyperpolarization. The range of molecules that can be hyperpolarized with PHIP significantly expanded in 2015 when PHIP by means of side arm hydrogenation (PHIP-SAH) was introduced. Herein, parahydrogen is added to an unsaturated alcoholic moiety of an ester followed by polarization transfer to carboxylate 13C nuclei with a subsequent side arm cleavage. In this review, the recent advances in PHIP-SAH are discussed, including the synthetic methodology to produce isotopically labeled precursors, peculiarities of pairwise addition of parahydrogen to PHIP-SAH precursors, polarization transfer approaches, hyperpolarization lifetime, side arm cleavage, purification of hyperpolarized solution, and, finally, in vitro and in vivo applications.
Collapse
Affiliation(s)
- Oleg G Salnikov
- Laboratory of Magnetic Resonance Microimaging, International Tomography Center SB RAS, 3A Institutskaya St., Novosibirsk, 630090, Russia
| | - Nikita V Chukanov
- Laboratory of Magnetic Resonance Microimaging, International Tomography Center SB RAS, 3A Institutskaya St., Novosibirsk, 630090, Russia
| | - Andrey N Pravdivtsev
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein and Kiel University, 24118, Kiel, Germany
| | - Dudari B Burueva
- Laboratory of Magnetic Resonance Microimaging, International Tomography Center SB RAS, 3A Institutskaya St., Novosibirsk, 630090, Russia
| | - Sergey V Sviyazov
- Laboratory of Magnetic Resonance Microimaging, International Tomography Center SB RAS, 3A Institutskaya St., Novosibirsk, 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia
| | - Kolja Them
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein and Kiel University, 24118, Kiel, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, Molecular Imaging North Competence Center (MOIN CC), Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein and Kiel University, 24118, Kiel, Germany
| | - Igor V Koptyug
- Laboratory of Magnetic Resonance Microimaging, International Tomography Center SB RAS, 3A Institutskaya St., Novosibirsk, 630090, Russia
| |
Collapse
|
70
|
Zhang J, Zhang S, Yu F, Wan Y, Wu M, Huang C. Unspliced XBP1 enhences metabolic reprogramming in colorectal cancer cells by interfering with the mitochondrial localization of MGME1. Biochem Biophys Res Commun 2025; 757:151613. [PMID: 40088676 DOI: 10.1016/j.bbrc.2025.151613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Tumor cells undergo metabolic reprogramming, which makes them tend to utilize anaerobic glycolysis rather than oxidation to rapidly produce energy and intermediate products required for proliferation. In this process, mitochondria inevitably undergo corresponding alterations; however, the specific alterations in mitochondria across different cancer types and the mechanisms governing these changes remain poorly understood. This study demonstrated that unspliced X-box binding protein 1 (XBP1-u) inhibits the translocation of mitochondrial genome maintenance exonuclease 1 (MGME1) into mitochondria by binding to the mitochondrial targeting sequence (MTS) of MGME1. This interaction results in the accumulation of mitochondrial 7sDNA, a reduction in mitochondrial DNA copy number, and a decrease in mitochondrial abundance. Consequently, this shift enhances the production of glycolysis and pentose phosphate pathway intermediates, thereby promoting the proliferation of colorectal cancer (CRC) cells. Our findings elucidated the critical mechanism by which XBP1-u enhances metabolic reprogramming by modulating mitochondrial biogenesis, and uncovered a novel role of MGME1 in the progression of CRC.
Collapse
Affiliation(s)
- Jiandong Zhang
- Metabolic Disease Research Center, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Suyang Zhang
- Metabolic Disease Research Center, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Fei Yu
- Metabolic Disease Research Center, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Yuting Wan
- Metabolic Disease Research Center, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Mingyue Wu
- Stomatologic Hospital&College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China.
| | - Can Huang
- Metabolic Disease Research Center, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
71
|
Karimpur-Zahmatkesh A, Khalaj-Kondori M. The perspective of targeting cancer cell metabolism: combination therapy approaches. Mol Biol Rep 2025; 52:375. [PMID: 40202553 DOI: 10.1007/s11033-025-10472-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells are considered the most adaptable for their metabolic status, which supports growth, survival, rapid proliferation, invasiveness, and metastasis in a nutrient-deficient microenvironment. Since the discovery of altered glucose metabolism (aerobic glycolysis), which is generally known as a part of metabolic reprogramming and an innate trait of cancer cells, in 1930 via Dr. Otto Warburg, numerous studies have endeavored to recognize various aspects of cancer cell metabolism and find new methods for efficiently eradicating described cells by targeting their energy metabolism. In this way, the outcomes have mainly been promising. Accordingly, outlining the related results will indeed assist us in making a definitive path for developing targeted therapy strategies based on cancer cell-altered metabolism. The present study reviews the key features of cancer cell metabolism and treatment strategies based on them. It emphasizes the importance of targeting cancer cell dysregulated metabolic pathways that influence the cell energy supply and manage cancer cell growth and survival. This trial also introduces a multimodal therapeutic strategy hypothesis, a potential next-generation combination therapy approach, and suggests interdisciplinary research to recognize the complexities of cancer metabolism and exploit them for designing more efficacious cancer therapeutic strategies.
Collapse
Affiliation(s)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
72
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
73
|
Franciosa G, Nieddu V, Battistini C, Caffarini M, Lupia M, Colombo N, Fusco N, Olsen JV, Cavallaro U. Quantitative Proteomics and Phosphoproteomics Analysis of Patient-Derived Ovarian Cancer Stem Cells. Mol Cell Proteomics 2025; 24:100965. [PMID: 40204276 DOI: 10.1016/j.mcpro.2025.100965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the deadliest gynecologic cancer. Key to the progression and ultimate lethality of this subtype is the intra-tumoral heterogeneity, which is defined as the coexistence of different cell types and populations within a single tumor. Among those, ovarian cancer stem cells (OCSCs) are a distinct subpopulation of tumor cells endowed with stem-like properties, which can survive current standard therapies, resulting in tumor recurrence. Here, we generated ex vivo primary OCSC-enriched three-dimensional (3D) spheres from 10 distinct treatment naive patient-derived adherent (2D) cultures. We used state-of-the-art quantitative mass spectrometry to characterize the molecular events associated with OCSCs by analyzing their proteome and phosphoproteome. Our data revealed a stemness-related protein signature, shared within a heterogeneous patient cohort, which correlates with chemo-refractoriness in a clinical proteomics dataset. Moreover, we identified targetable deregulated kinases and aberrant PDGF receptor activation in OCSCs. Pharmacological inhibition of PDGFR in adherent OC cells reduced the stemness potential, measured by sphere formation assay. Overall, we provide a valuable resource to identify new OCSC markers and putative targets for OCSC-directed therapies.
Collapse
Affiliation(s)
- Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular andMolecular Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark.
| | - Valentina Nieddu
- Unit of Gynecological Oncology Research, European Institute of Oncology IRCSS, Milano, Italy
| | - Chiara Battistini
- Unit of Gynecological Oncology Research, European Institute of Oncology IRCSS, Milano, Italy
| | - Miriam Caffarini
- Unit of Gynecological Oncology Research, European Institute of Oncology IRCSS, Milano, Italy
| | - Michela Lupia
- Unit of Gynecological Oncology Research, European Institute of Oncology IRCSS, Milano, Italy
| | - Nicoletta Colombo
- Division of Gynecologic Oncology, European Institute of Oncology IRCCS, Milano, Italy; Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Nicola Fusco
- Department of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milano, Italy
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular andMolecular Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark.
| | - Ugo Cavallaro
- Unit of Gynecological Oncology Research, European Institute of Oncology IRCSS, Milano, Italy.
| |
Collapse
|
74
|
Park SJ, Cerella C, Kang JM, Byun J, Kum D, Orlikova-Boyer B, Lorant A, Schnekenburger M, Al-Mourabit A, Christov C, Lee J, Han BW, Diederich M. Tetrahydrobenzimidazole TMQ0153 targets OPA1 and restores drug sensitivity in AML via ROS-induced mitochondrial metabolic reprogramming. J Exp Clin Cancer Res 2025; 44:114. [PMID: 40197337 PMCID: PMC11974110 DOI: 10.1186/s13046-025-03372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly aggressive cancer with a 5-year survival rate of less than 35%. It is characterized by significant drug resistance and abnormal energy metabolism. Mitochondrial dynamics and metabolism are crucial for AML cell survival. Mitochondrial fusion protein optic atrophy (OPA)1 is upregulated in AML patients with adverse mutations and correlates with poor prognosis. METHOD This study investigated targeting OPA1 with TMQ0153, a tetrahydrobenzimidazole derivative, to disrupt mitochondrial metabolism and dynamics as a novel therapeutic approach to overcome treatment resistance. Effects of TMQ0153 treatment on OPA1 and mitofusin (MFN)2 protein levels, mitochondrial morphology, and function in AML cells. In this study, we examined reactive oxygen species (ROS) production, oxidative phosphorylation (OXPHOS) inhibition, mitochondrial membrane potential (MMP) depolarization, and apoptosis. Additionally, metabolic profiling was conducted to analyze changes in metabolic pathways. RESULTS TMQ0153 treatment significantly reduced OPA1 and mitofusin (MFN)2 protein levels and disrupted the mitochondrial morphology and function in AML cells. This increases ROS production and inhibits OXPHOS, MMP depolarization, and caspase-dependent apoptosis. Metabolic reprogramming was observed, shifting from mitochondrial respiration to glycolysis and impaired respiratory chain activity. Profiling revealed reduced overall metabolism along with changes in the glutathione (GSH)/oxidized glutathione (GSSG) and NAD⁺/NADH redox ratios. TMQ0153 treatment reduces tumor volume and weight in MV4-11 xenografts in vivo. Combination therapies with TMQ0153 and other AML drugs significantly reduced the leukemic burden and prolonged survival in NOD scid gamma (NSG) mice xenografted with U937-luc and MOLM-14-luc cells. CONCLUSION TMQ0153 targets mitochondrial dynamics by inhibiting OPA1, inducing metabolic reprogramming, and triggering apoptosis in AML cells. It enhances the efficacy of existing AML therapies and provides a promising combination treatment approach that exploits mitochondrial vulnerability and metabolic reprogramming to improve treatment outcomes in AML.
Collapse
MESH Headings
- Humans
- GTP Phosphohydrolases/metabolism
- GTP Phosphohydrolases/antagonists & inhibitors
- GTP Phosphohydrolases/genetics
- Reactive Oxygen Species/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Animals
- Mice
- Benzimidazoles/pharmacology
- Mitochondria/metabolism
- Mitochondria/drug effects
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Apoptosis/drug effects
- Oxidative Phosphorylation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Metabolic Reprogramming
Collapse
Affiliation(s)
- Su Jung Park
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jin Mo Kang
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinyoung Byun
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - David Kum
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Barbora Orlikova-Boyer
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Anne Lorant
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Luxembourg Centre for Systems Biomedicine, Bioinformatics Core, Roudeneck, 1, Boulevard du Jazz, Esch-sur-Alzette, L-4370, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Ali Al-Mourabit
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-Sur-Yvette, 91190, France
| | - Christo Christov
- Service d'Histologie, Faculté de Médicine, Université de Lorraine, and INSERM U1256 NGERE, 54000, Nancy, France
| | - Juyong Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Marc Diederich
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
75
|
Dumas L, Shin S, Rigaud Q, Cargnello M, Hernández-Suárez B, Herviou P, Saint-Laurent N, Leduc M, Le Gall M, Monchaud D, Dassi E, Cammas A, Millevoi S. RNA G-quadruplexes control mitochondria-localized mRNA translation and energy metabolism. Nat Commun 2025; 16:3292. [PMID: 40195294 PMCID: PMC11977240 DOI: 10.1038/s41467-025-58118-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
Cancer cells rely on mitochondria for their bioenergetic supply and macromolecule synthesis. Central to mitochondrial function is the regulation of mitochondrial protein synthesis, which primarily depends on the cytoplasmic translation of nuclear-encoded mitochondrial mRNAs whose protein products are imported into mitochondria. Despite the growing evidence that mitochondrial protein synthesis contributes to the onset and progression of cancer, and can thus offer new opportunities for cancer therapy, knowledge of the underlying molecular mechanisms remains limited. Here, we show that RNA G-quadruplexes (RG4s) regulate mitochondrial function by modulating cytoplasmic mRNA translation of nuclear-encoded mitochondrial proteins. Our data support a model whereby the RG4 folding dynamics, under the control of oncogenic signaling and modulated by small molecule ligands or RG4-binding proteins, modifies mitochondria-localized cytoplasmic protein synthesis. Ultimately, this impairs mitochondrial functions, affecting energy metabolism and consequently cancer cell proliferation.
Collapse
Affiliation(s)
- Leïla Dumas
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Sauyeun Shin
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Quentin Rigaud
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Marie Cargnello
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Beatriz Hernández-Suárez
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Pauline Herviou
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Nathalie Saint-Laurent
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Marjorie Leduc
- Proteom'IC facility, Université Paris Cité, CNRS, INSERM Institut Cochin, Paris, France
| | - Morgane Le Gall
- Proteom'IC facility, Université Paris Cité, CNRS, INSERM Institut Cochin, Paris, France
| | - David Monchaud
- Institut de Chimie Moléculaire (ICMUB), UBFC Dijon CNRS UMR6302, Dijon, France
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, TN, Italy.
| | - Anne Cammas
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France.
| | - Stefania Millevoi
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Equipe Labellisée Fondation ARC, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France.
| |
Collapse
|
76
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
77
|
Gentile R, Feudi D, Sallicandro L, Biagini A. Can the Tumor Microenvironment Alter Ion Channels? Unraveling Their Role in Cancer. Cancers (Basel) 2025; 17:1244. [PMID: 40227837 PMCID: PMC11988140 DOI: 10.3390/cancers17071244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Neoplastic cells are characterized by metabolic reprogramming, known as the Warburg effect, in which glucose metabolism is predominantly directed toward aerobic glycolysis, with reduced mitochondrial oxidative phosphorylation and increased lactate production even in the presence of oxygen. This phenomenon provides cancer cells with a proliferative advantage, allowing them to rapidly produce energy (in the form of ATP) and generate metabolic intermediates necessary for the biosynthesis of macromolecules essential for cell growth. It is important to understand the role of ion channels in the tumor context since they participate in various physiological processes and in the regulation of the tumor microenvironment. These changes may contribute to the development and transformation of cancer cells, as well as affect the communication between cells and the surrounding microenvironment, including impaired or altered expression and functionality of ion channels. Therefore, the aim of this review is to elucidate the impact of the tumor microenvironment on the electrical properties of the cellular membranes in several cancers as a possible therapeutic target.
Collapse
Affiliation(s)
- Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
| | - Davide Feudi
- Department of Biostatistics, Epidemiology and Public Health, University of Padua, Via L. Loredan 18, 35131 Padova, Italy;
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| |
Collapse
|
78
|
Nikopaschou M, Samiotaki M, Stylianaki EA, Król K, Gragera P, Raja A, Aidinis V, Chroni A, Fruci D, Panayotou G, Stratikos E. ERAP1 Activity Modulates the Immunopeptidome but Also Affects the Proteome, Metabolism, and Stress Responses in Cancer Cells. Mol Cell Proteomics 2025; 24:100964. [PMID: 40189142 DOI: 10.1016/j.mcpro.2025.100964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
Endoplasmic reticulum (ER) aminopeptidase 1 (ERAP1) metabolizes peptides inside the ER and shapes the peptide repertoire available for binding to major histocompatibility complex class I molecules (MHC-I). However, it may have additional effects on cellular homeostasis, which have not been explored. To address these questions, we used both genetic silencing of ERAP1 expression as well as treatment with a selective allosteric ERAP1 inhibitor to probe changes in the immunopeptidome and proteome of the A375 melanoma cancer cell line. We observed significant immunopeptidome shifts with both methods of functional ERAP1 disruption, which were distinct for each method. Both methods of inhibition led to an enhancement, albeit slight, in tumor cell killing by stimulated human peripheral blood mononuclear cells and in significant proteomic alterations in pathways related to metabolism and cellular stress. Similar proteomic changes were also observed in the leukemia cell line THP-1. Biochemical analyses suggested that ERAP1 inhibition affected sensitivity to ER stress, reactive oxygen species production, and mitochondrial metabolism. Although the proteomics shifts were significant, their potential in shaping immunopeptidome shifts was limited since only 9.6% of differentially presented peptides belonged to proteins with altered expression and only 4.0% of proteins with altered expression were represented in the immunopeptidome shifts. Taken together, our findings suggest that modulation of ERAP1 activity can generate unique immunopeptidomes, mainly due to altered peptide processing in the ER, but also induce changes in the cellular proteome and metabolic state which may have further effects on tumor cells.
Collapse
Affiliation(s)
- Martha Nikopaschou
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece; Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Greece
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Bioinnovation, Vari, Greece
| | - Elli-Anna Stylianaki
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Fundamental Biomedical Research, Vari, Greece
| | - Kamila Król
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paula Gragera
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Aroosha Raja
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Vassilis Aidinis
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Fundamental Biomedical Research, Vari, Greece
| | - Angeliki Chroni
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece
| | - Doriana Fruci
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - George Panayotou
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Bioinnovation, Vari, Greece
| | - Efstratios Stratikos
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece; Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Greece.
| |
Collapse
|
79
|
Somlyai G, Papp A, Somlyai I, Kovács BZ, Debrődi M. Real-World Data Confirm That the Integration of Deuterium Depletion into Conventional Cancer Therapy Multiplies the Survival Probability of Patients. Biomedicines 2025; 13:876. [PMID: 40299476 PMCID: PMC12025113 DOI: 10.3390/biomedicines13040876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Over thirty years of basic research has demonstrated that the deuterium-to-hydrogen ratio plays a pivotal role in regulating metabolism and cell growth via a sub-molecular regulatory system that orchestrates the intricate complexity of life in eukaryotic organisms. Deuterium depletion, achieved through deuterium-depleted water (DDW), has shown anticancer effects in vitro, in vivo, and in Phase 2 prospective and retrospective clinical studies. Methods: In this population-based observational study, 2649 cancer patients undergoing conventional therapy and consuming DDW were included between October 1992 and October 2024. With various cancer types and stages and conventional therapies received, they are representing a broad spectrum of the Hungarian cancer population. Survival was selected as the primary endpoint, and the median survival time (MST) of these patients and various subgroups was calculated and compared to the overall Hungarian cancer population's MST of 2.4 years. Results: For the entire study population, MST from diagnosis was 12.4 years (95% CI: 9.8-14.9), and from the initiation of DDW treatment, 7.6 years (95% CI: 5.9-9.3). Conclusions: Utilizing DDW enables targeted intervention in the sub-molecular regulatory system, paving the way for innovative therapeutic applications and a more profound understanding of cellular processes. Integrating deuterium depletion into conventional cancer therapies has the potential to significantly enhance survival rates and reduce cancer-related mortality by 75-80%.
Collapse
Affiliation(s)
- Gábor Somlyai
- HYD LLC for Cancer Research and Drug Development, Villányi út 97, 1118 Budapest, Hungary (B.Z.K.); (M.D.)
| | - András Papp
- Department of Public Health, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Ildikó Somlyai
- HYD LLC for Cancer Research and Drug Development, Villányi út 97, 1118 Budapest, Hungary (B.Z.K.); (M.D.)
| | - Beáta Zs Kovács
- HYD LLC for Cancer Research and Drug Development, Villányi út 97, 1118 Budapest, Hungary (B.Z.K.); (M.D.)
| | - Mária Debrődi
- HYD LLC for Cancer Research and Drug Development, Villányi út 97, 1118 Budapest, Hungary (B.Z.K.); (M.D.)
| |
Collapse
|
80
|
Nangia-Makker P, Ahrens M, Purandare N, Aras S, Li J, Gurdziel K, Jang H, Kim S, Shekhar MP. Relationship between melanoma vemurafenib tolerance thresholds and metabolic pathway choice and Wnt signaling involvement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641924. [PMID: 40093038 PMCID: PMC11908245 DOI: 10.1101/2025.03.06.641924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Vemurafenib constitutes an important therapeutic for BRAFV600 mutant melanomas, but despite high initial response rates, resistance to BRAF and MEK inhibitors quickly develops. Here, we performed an integrative analysis of metabolomic consequences and transcriptome alterations to uncover mechanisms involved in adaptive vemurafenib resistance (VemR) development and their relationship with vemurafenib tolerance thresholds. We developed BRAFV600E isogenic models of VemR utilizing M14 and A2058 lines, and patient-derived melanomas with V600E or normal BRAF to verify vemurafenib selectivity. MEK or PI3K inhibitors only partially inhibited VemR cell proliferation, indicating cross-resistance to these inhibitors. MITF and β-catenin levels were induced and treatment with Wnt/β-catenin inhibitor ICG-001 restored vemurafenib sensitivity with concomitant reductions in β-catenin-regulated gene expressions, phospho-ERK1/2, and VemR-induced mitochondrial mass and respiration. Targeted metabolite, MitoPlate-S1, Mito-stress and transcriptome/metabolomic analysis showed that melanoma cells with elevated vemurafenib tolerance thresholds such as A2058 VemR cells utilize Wnt/β-catenin signaling for mitochondrial metabolism while VemR cells with low tolerance such as M14 VemR cells rely on Wnt/β-catenin signaling for pentose phosphate pathway. Pathways associated with cytokine-cytokine receptor, ECM receptor, and neuroactive ligand receptor interactions were similarly enriched in BRAFV600E patient-derived melanoma as M14 and A2058 cells whereas distinct pathways involving cell cycle, DNA replication, Fanconi anemia and DNA repair pathways are upregulated in wild type BRAF expressing patient derived melanoma. These data show for the first time that the metabolic pathway choices made by VemR BRAF mutant melanomas are controlled by vemurafenib tolerance and endurance thresholds and Wnt/β-catenin signaling plays a central role in coordinating expression of genes controlling VemR and metabolic pathway shifts.
Collapse
|
81
|
McMorrow R, de Bruijn HS, Farina S, van Ardenne RJ, Que I, Mastroberardino PG, Robinson DJ, Mezzanotte L, Löwik CW. Combination of Bremachlorin PDT and Immune Checkpoint Inhibitor Anti-PD-1 Shows Response in Murine Immunological T-cell-High and T-cell-Low PDAC Models. Mol Cancer Ther 2025; 24:605-617. [PMID: 39704624 PMCID: PMC11962392 DOI: 10.1158/1535-7163.mct-23-0733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/12/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most challenging types of cancer with little or no response to immune checkpoint inhibitors (ICI). Photodynamic therapy (PDT) has been shown to ablate tumors and induce an immune response. In our study, we investigated the effect of PDT using the photosensitizer Bremachlorin, in its ability to reduce tumor burden and immunologically sensitize T-cell-high and T-cell-low murine PDAC tumors to the ICIs that blocks PD-1 immune checkpoint. In addition, we monitored the effect on survival and investigated if there was a response in PDT-treated and non-PDT-treated distant tumors. Our results showed that Bremachlorin PDT induces direct tumor killing that increased survival in both "hot" T-cell-high and "cold" T-cell-low PDAC tumors and that it can make T-cell-high tumors more sensitive to ICIs blocking PD-1. We found that T-cell-high tumor-bearing mice had an overall greater response to therapy than did T-cell-low tumor-bearing mice. One mouse with T-cell-high tumors exhibited complete tumor regression in both the treated and nontreated distant tumor 90 days after treatment. These results indicate that combining ICIs with Bremachlorin PDT could be a promising therapeutic intervention for enhancing PDAC's response to therapy.
Collapse
Affiliation(s)
- Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Henriette S. de Bruijn
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Stefania Farina
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ruben J.L. van Ardenne
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ivo Que
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
- IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Life, Health, and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Dominic J. Robinson
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Clemens W.G.M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
82
|
Trujillo MN, Jennings EQ, Farrera DO, Kitamura N, Anderson CC, Gehrke S, Reisz JA, Johannsen M, Roede JR, D'Alessandro A, Galligan JJ. Glyoxalase 2 Coordinates de Novo Serine Metabolism. Chembiochem 2025; 26:e202401086. [PMID: 39988553 DOI: 10.1002/cbic.202401086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
Phosphoglycerate dehydrogenase (PHGDH) is the first enzyme in de novo Ser biosynthesis. Numerous metabolic pathways rely on Ser as a precursor, most notably one-carbon metabolism, glutathione biosynthesis, and de novo nucleotide biosynthesis. To facilitate proliferation, many cancer cells shunt glycolytic flux through this pathway, placing PHGDH as a metabolic liability and feasible therapeutic target for the treatment of cancer. Herein, we demonstrate the post-translational modification (PTM) of PHGDH by lactoylLys. These PTMs are generated through a non-enzymatic acyl transfer from the glyoxalase cycle intermediate, lactoylglutathione (LGSH). Knockout of the primary LGSH regulatory enzyme, glyoxalase 2 (GLO2), results in increased LGSH and resulting lactoylLys modification of PHGDH. These PTMs reduce enzymatic activity, resulting in a marked reduction in intracellular Ser. Using stable isotope tracing, we demonstrate reduced flux through the de novo Ser biosynthetic pathway. Collectively, these data identify PHGDH as a target for modification by lactoylLys, resulting in reduced enzymatic activity and reduced intracellular Ser.
Collapse
Affiliation(s)
- Marissa N Trujillo
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, 85721, USA
| | - Erin Q Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, 85721, USA
| | - Dominique O Farrera
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, 85721, USA
| | - Naoya Kitamura
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, 85721, USA
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, 80045, USA
| | - Sarah Gehrke
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, 80045, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, 80045, USA
| | - Mogens Johannsen
- Department of Forensic Medicine, Aarhus University, 8200, Aarhus, Denmark
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, 80045, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, 85721, USA
| |
Collapse
|
83
|
Tomar MS, Mohit, Kumar A, Shrivastava A. Circadian immunometabolism: A future insight for targeted therapy in cancer. Sleep Med Rev 2025; 80:102031. [PMID: 39603026 DOI: 10.1016/j.smrv.2024.102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Circadian rhythms send messages to regulate the sleep-wake cycle in living beings, which, regulate various biological activities. It is well known that altered sleep-wake cycles affect host metabolism and significantly deregulate the host immunity. The dysregulation of circadian-related genes is critical for various malignancies. One of the hallmarks of cancer is altered metabolism, the effects of which spill into surrounding microenvironments. Here, we review the emerging literature linking the circadian immunometabolic axis to cancer. Small metabolites are the products of various metabolic pathways, that are usually perturbed in cancer. Genes that regulate circadian rhythms also regulate host metabolism and control metabolite content in the tumor microenvironment. Immune cell infiltration into the tumor site is critical to perform anticancer functions, and altered metabolite content affects their trafficking to the tumor site. A compromised immune response in the tumor microenvironment aids cancer cell proliferation and immune evasion, resulting in metastases. The role of circadian rhythms in these processes is largely overlooked and demands renewed attention in the search for targets against cancer growth and spread. The precision medicine approach requires targeting the circadian immune metabolism in cancer.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Mohit
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India; Department of Prosthodontics, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India.
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India.
| |
Collapse
|
84
|
Gnocchi D, Nikolic D, Russo S, Matrella ML, Paparella RR, Kumar S, Karki SS, Sabbà C, Cocco T, Lobasso S, Mazzocca A. Dysfunctional mitochondrial bioenergetics sustains drug resistance in cancer cells. Am J Physiol Cell Physiol 2025; 328:C1150-C1159. [PMID: 39853268 DOI: 10.1152/ajpcell.00538.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/20/2024] [Accepted: 01/16/2025] [Indexed: 01/26/2025]
Abstract
Resistance to drugs is one of the major issues affecting the response to pharmacological treatments for tumors. Different mechanisms have been proposed to explain the development of cancer drug resistance (CDR), and several approaches to overcome it have been suggested. However, the biological basis of CDR remains unclear. Here, we investigated whether mitochondrial damage and consequent mitochondrial dysfunction are major causes of drug resistance in different tumors. To this end, we used cell lines from three tumors: hepatocellular carcinoma, breast cancer, and colon cancer. We then applied a protocol that recapitulates chemotherapy regimens in patients, rendering each cell line resistant to the drug commonly used in their respective treatments. The combination of cellular respiration analysis, gene expression analysis of cytochrome c oxidase isoforms, and mass spectrometry assessment of cardiolipin (CL) reveals that mitochondrial dysfunction is the underlying cause of the resistant phenotype. Importantly, we disclosed for the first time the rapid inhibition of oxidative phosphorylation (OXPHOS) by l-lactate, the major product of fermentation. Finally, we demonstrated that inhibition of lactic acid fermentation and activation of OXPHOS can increase drug sensitivity in all tested drug-resistant cancer cells. Taken together, our results suggest that inhibiting fermentation and enhancing mitochondrial function in cancer cells may be a concrete option to control the worrisome phenomenon of CDR.NEW & NOTEWORTHY Cancer drug resistance (CDR) is increasingly becoming a concerning clinical problem. The mechanisms behind the onset of CDR are still not well defined. In this study, we demonstrated that a treatment mimicking long-term clinical protocols with commonly used chemotherapeutic agents promotes mitochondrial bioenergetic dysfunction, leading to the acquisition of CDR. In a future perspective, interventions aimed at inhibiting fermentation and restoring OXPHOS efficiency may offer tangible opportunities to reduce the clinical burden of CDR.
Collapse
Affiliation(s)
- Davide Gnocchi
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro School of Medicine, Piazza G. Cesare, Bari, Italy
| | - Dragana Nikolic
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro School of Medicine, Piazza G. Cesare, Bari, Italy
| | - Silvia Russo
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro School of Medicine, Bari, Italy
| | - Maria Laura Matrella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro School of Medicine, Bari, Italy
| | - Rosa R Paparella
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro School of Medicine, Piazza G. Cesare, Bari, Italy
| | - Sujeet Kumar
- Department of Pharmaceutical Chemistry, Dr Prabhakar B Kore Basic Science Research Center, Off-campus, KLE College of Pharmacy, (A Constituent Unit of KLE Academy of Higher Education and Research, Belagai), Bengaluru, India
| | - Subhas S Karki
- Department of Pharmaceutical Chemistry, Dr Prabhakar B Kore Basic Science Research Center, Off-campus, KLE College of Pharmacy, (A Constituent Unit of KLE Academy of Higher Education and Research, Belagai), Bengaluru, India
| | - Carlo Sabbà
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro School of Medicine, Piazza G. Cesare, Bari, Italy
| | - Tiziana Cocco
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro School of Medicine, Bari, Italy
| | - Simona Lobasso
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro School of Medicine, Bari, Italy
| | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro School of Medicine, Piazza G. Cesare, Bari, Italy
| |
Collapse
|
85
|
Charlot A, Bringolf A, Mallard J, Jaulin A, Crouchet E, Charles AL, Duteil D, Alpy F, Tomasetto CL, Baumert TF, Zoll J. Mammary tumor development induces perturbation of liver glucose metabolism with inflammation and fibrosis. Am J Physiol Endocrinol Metab 2025; 328:E645-E651. [PMID: 40106327 DOI: 10.1152/ajpendo.00498.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/01/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Cancer cells rely on glycolysis and lactic fermentation for ATP production, inducing an abnormal glucose uptake in tumors. However, it is largely unknown whether the increased tumor glucose consumption affects overall body glucose homeostasis including perturbation of the liver glucose production pathways. The effect of mammary tumor development on the liver metabolism pathway was examined by using a mouse model based on FVB/N wild-type (WT-SD) and FVB/N-Tg(MMTV-PyVT)634Mul/J mice (Tg-SD), who develop spontaneous mammary tumors. Blood and livers were analyzed for metabolic changes, by measuring histological staining, signaling, and insulin sensitivity. Tg-SD mice developed mammary tumors with an average weight of 6 g, and cancer development increased total food intake without impacting body weight gain. Tumor development did not affect blood glycemia and lactate levels but increased insulin and homeostasis model assessment of insulin resistance (HOMA-IR) index (P = 0.06). In the liver, Tg-SD mice with tumors exhibited a decrease in glycogen content and an increase in gluconeogenesis gene expression, as G6pc, Pgc1α, and Foxo1 (P < 0.05), as well as Pepck and Ldha (P < 0.01). Moreover, the phosphorylation of AMPK and AKT was significantly decreased (P < 0.01 and P < 0.05, respectively). Surprisingly, liver fibrosis was markedly increased in Tg mice (P < 0.05) alongside elevated inflammatory gene expression, such as IL1β (P < 0.01) or IL6 (P < 0.05). Here, we found that the development of non-metastatic mammary tumors using the MMTV-PyMT mouse model disrupts liver function through the development of inflammation, fibrosis, and metabolic perturbation, including an increase in glucose production and insulin resistance. Finally, these observations unravel a previously unknown metabolic cross talk between the tumors and the liver.NEW & NOTEWORTHY This work demonstrates that the spontaneous development of non-metastatic mammary tumors triggers hepatic activation of endogenous glucose production pathways, coinciding with the onset of insulin resistance. This finding suggests a significant cross talk between tumors and the liver during tumorigenesis, aiming at enhancing glucose production to meet the elevated energy demands of the tumor. Understanding this interaction could provide insights into metabolic alterations associated with cancer and lead to potential therapeutic targets to inhibit tumor metabolism.
Collapse
Affiliation(s)
- Anouk Charlot
- Institute for Translational Medicine and Liver Disease, UMR-S1110, Inserm, University of Strasbourg, Strasbourg, France
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072 "Mitochondrie, Stress oxydant et Plasticité musculaire,"University of Strasbourg, Strasbourg, France
| | - Anthony Bringolf
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072 "Mitochondrie, Stress oxydant et Plasticité musculaire,"University of Strasbourg, Strasbourg, France
| | - Joris Mallard
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072 "Mitochondrie, Stress oxydant et Plasticité musculaire,"University of Strasbourg, Strasbourg, France
- Faculty of Sport Sciences, University of Strasbourg, Strasbourg, France
- Institute of Cancerology Strasbourg Europe (ICANS), Strasbourg, France
| | - Amélie Jaulin
- IGBMC UMR 7104-UMR-S 1258, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Emilie Crouchet
- Institute for Translational Medicine and Liver Disease, UMR-S1110, Inserm, University of Strasbourg, Strasbourg, France
| | - Anne-Laure Charles
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072 "Mitochondrie, Stress oxydant et Plasticité musculaire,"University of Strasbourg, Strasbourg, France
| | - Delphine Duteil
- IGBMC UMR 7104-UMR-S 1258, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | - Fabien Alpy
- IGBMC UMR 7104-UMR-S 1258, CNRS, Inserm, Université de Strasbourg, Illkirch, France
| | | | - Thomas F Baumert
- Institute for Translational Medicine and Liver Disease, UMR-S1110, Inserm, University of Strasbourg, Strasbourg, France
- IHU Strasbourg, Strasbourg, France
- Service d'Hépato-gastroentérologie, University Hospital of Strasbourg, Strasbourg, France
| | - Joffrey Zoll
- Biomedicine Research Center of Strasbourg (CRBS), UR 3072 "Mitochondrie, Stress oxydant et Plasticité musculaire,"University of Strasbourg, Strasbourg, France
- Service de Physiologie et Explorations Fonctionnelles, University Hospital of Strasbourg, Strasbourg, France
- Faculty of Medicine, University of Strasbourg, Strasbourg, France
| |
Collapse
|
86
|
Gao D, Yam JWP. Reversal of lipid metabolism as a therapeutic approach for HCC. Hepatology 2025; 81:1113-1115. [PMID: 38935863 DOI: 10.1097/hep.0000000000001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Affiliation(s)
- Daming Gao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| |
Collapse
|
87
|
Gao F, Shah R, Xin G, Wang R. Metabolic Dialogue Shapes Immune Response in the Tumor Microenvironment. Eur J Immunol 2025; 55:e202451102. [PMID: 40223597 PMCID: PMC11995254 DOI: 10.1002/eji.202451102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025]
Abstract
The fate of immune cells is fundamentally linked to their metabolic program, which is also influenced by the metabolic landscape of their environment. The tumor microenvironment represents a unique system for intercellular metabolic interactions, where tumor-derived metabolites suppress effector CD8+ T cells and promote tumor-promoting macrophages, reinforcing an immune-suppressive niche. This review will discuss recent advancements in metabolism research, exploring the interplay between various metabolites and their effects on immune cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Fengxia Gao
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Rushil Shah
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| | - Gang Xin
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Ruoning Wang
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
88
|
Sharma S, Sagar R. Efficient synthesis of coumarin based triazole linked O-glycoconjugates as new bio-active glycohybrids. Carbohydr Res 2025; 550:109395. [PMID: 39864121 DOI: 10.1016/j.carres.2025.109395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
Glycohybrids are biologically significant molecules with variety of biological functions and are found as structural motifs in numerous natural products. Here, we report the synthesis of various new coumarin-based O-glycoconjugates as glycohybrids that are chirally enriched and bridged by 1,2,3-triazoles ring system. The1,2,3-triazoles bridging was done via CuAAC click-chemistry. Click chemistry offers several advantages, including high chemo- and regioselectivity, mild reaction conditions, easy purification, and compatibility with multiple functional groups. Two series of O-glycoconjugates as new glycohybrids were designed and efficiently synthesized in very good isolated yields, using d-glucose, d-galactose, d-mannose, d-arabinose, 3,4,6-tri-O-acetyl-D-glucal, 3,4,6-tri-O-acetyl-D-galactal and 3,4-di-O-acetyl-D-arabinal derived 1-O-propargylated glycosides, reacting with various 4-azidocoumarins under click-chemistry reaction conditions. The prepared new coumarin-based O-glycoconjugates as glycohybrids were found to possess anticancer activity against MCF-7 breast cancer cell lines at micromolar concentration.
Collapse
Affiliation(s)
- Sunil Sharma
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ram Sagar
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
89
|
Jung JH, Yang Y, Kim Y. Hypoxia‑induced SREBP1‑mediated lipogenesis and autophagy promote cell survival via fatty acid oxidation in breast cancer cells. Oncol Lett 2025; 29:175. [PMID: 39975955 PMCID: PMC11837466 DOI: 10.3892/ol.2025.14921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025] Open
Abstract
In the hypoxic tumor microenvironment, cancer cells undergo metabolic reprogramming to survive. The present study aimed to assess the effects of hypoxic conditions on the lipid metabolism of breast cancer cells to elucidate the mechanisms by which cancer cells survive in an unfavorable environment. Cell viability was assessed by trypan blue staining, MTT and Annexin V-PI assays. Intracellular lipid levels were quantified using Nile red stain with immunofluorescence (IF). Autophagy was detected using LC3 antibody, Cyto-ID stain, IF, Western blotting, and flow cytometry. Fatty acid oxidation (FAO) and ATP production were analyzed using specific assays, while gene expression was assessed by reverse transcription-polymerase chain reaction. siRNA transfection was used for gene knockdown, and Kaplan-Meier analysis was performed for survival analysis. Fatostatin and rapamycin served as an inhibitor of sterol regulatory element-binding protein 1 (SREBP1) and an autophagy inducer, respectively. Under hypoxic conditions, triple-negative breast cancer (TNBC) MDA-MB-231 cells showed markedly increased survival and proliferation rates compared with normal cells (MCF-10A) and estrogen receptor-positive cells (MCF-7), with no change in apoptosis. Under hypoxic conditions, MDA-MB-231 cells showed increased expression of lipogenesis, autophagy and FAO-related enzymes and activation of SREBP1, a key transcription factor for lipogenic genes, whereas these changes were not observed in MCF-7 cells. When SREBP1 was inhibited with chemical inhibitors and siRNA, the expression of lipogenic, autophagic and FAO-related enzymes decreased, resulting in reduced ATP production and viability in hypoxic MDA-MB-231 cells; however, this effect was restored when an autophagy inducer was added. Kaplan-Meier analysis demonstrated that higher SREBP1 expression in patients with TNBC was associated with a worse prognosis, suggesting that SREBP1-mediated reprogramming of lipid metabolism and autophagy under hypoxia is essential for TNBC cell survival. The results of the present study indicate that strategies targeting SREBP1 could be exploited to treat TNBC and improve prognosis.
Collapse
Affiliation(s)
- Jae-Ha Jung
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- BK 21 FOUR Program for Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeseul Yang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Yongbaek Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
90
|
Hayashi Y, Saeki A, Yoshimoto S, Yano E, Yasukochi A, Kimura S, Utsunomiya T, Minami K, Aso Y, Hatakeyama Y, Lo YC, Hirata M, Jimi E, Kawakubo-Yasukochi T. 4-Octyl Itaconate Attenuates Cell Proliferation by Cellular Senescence via Glutathione Metabolism Disorders and Mitochondrial Dysfunction in Melanoma. Antioxid Redox Signal 2025; 42:547-565. [PMID: 39931827 DOI: 10.1089/ars.2024.0629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Aims: Itaconate (IA) is synthesized in the citric acid cycle via cis-aconitate decarboxylase (ACOD1); however, its biological significance in cancer remains incompletely understood. In previous studies, 4-octyl itaconate (OI) was used as a membrane-permeable form of IA, but little detailed verification of the difference in biological activities between IA and OI exists. Here, we investigated the direct effects of IA and OI on melanoma. Results: The proliferation of melanoma cells treated with OI was significantly suppressed in vitro, and our transcriptomic analysis revealed drastic changes in the expression of glutathione metabolism-related genes in OI-treated cells. Indeed, OI treatment decreased intracellular glutathione levels, followed by increased production of reactive oxygen species and expression of γH2AX, a marker of DNA damage, and β-galactosidase, a marker of cellular senescence. We further showed that the mitochondrial respiratory capacity in B16 cells was significantly decreased by OI treatment. OI administration also suppressed the growth of B16 tumor transplants in vivo, and the expression of γH2AX was increased in tumor tissues of OI-treated mice. In addition, minimal effects of OI treatment were observed in melanocytes and normal tissues. We also proved that not only exogenous IA, which enters intracellularly, but also endogenous IA has little effect on melanoma proliferation activity, via an investigation using Acod1-overexpressing transfectants and Acod1-deficient mice. Conclusion: This work revealed that OI disrupts the antioxidant system via the collapse of glutathione metabolism and inhibits cancer cell proliferation. Antioxid. Redox Signal. 42, 547-565.
Collapse
Affiliation(s)
- Yoshikazu Hayashi
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Ayaka Saeki
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shohei Yoshimoto
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
- Division of Biomedical Sciences, Department of Morphological Biology, Section of Pathology, Fukuoka Dental College, Fukuoka, Japan
| | - Ena Yano
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Atsushi Yasukochi
- Division of Maxillofacial Diagnostic and Surgical Sciences, Section of Oral and Maxillofacial Oncology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Soi Kimura
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomoe Utsunomiya
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Kento Minami
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuji Aso
- Department of Biobased Materials Science, Kyoto Institute of Technology, Kyoto, Japan
| | - Yuji Hatakeyama
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Yi-Chen Lo
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Eijiro Jimi
- OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
91
|
Sobti A, Skinner H, Wilke CT. Predictors of Radiation Resistance and Novel Radiation Sensitizers in Head and Neck Cancers: Advancing Radiotherapy Efficacy. Semin Radiat Oncol 2025; 35:224-242. [PMID: 40090749 DOI: 10.1016/j.semradonc.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/18/2025]
Abstract
Radiation resistance in head and neck squamous cell carcinoma (HNSCC), driven by intrinsic and extrinsic factors, poses a significant challenge in radiation oncology. The key contributors are tumor hypoxia, cancer stem cells, cell cycle checkpoint activation, and DNA repair processes (homologous recombination and non-homologous end-joining). Genetic modifications such as TP53 mutations, KRAS mutations, EGFR overexpression, and abnormalities in DNA repair proteins like BRCA1/2 additionally affect radiation sensitivity. Novel radiosensitizers targeting these pathways demonstrate the potential to overcome resistance. Hypoxia-activated drugs and gold nanoparticles enhance the efficacy of radiotherapy and facilitate targeted distribution. Integrating immunotherapy, especially immune checkpoint inhibitors, with radiation therapy, enhances anti-tumor responses and reduces resistance. Epigenetic alterations, such as DNA methylation and histone acetylation, significantly influence radiation response, with the potential for sensitization through histone deacetylase inhibitors and non-coding RNA regulators. Metabolic changes linked to glucose, lipid, and glutamine metabolism influence radiosensitivity, uncovering new targets for radiosensitization. Human papillomavirus (HPV)-associated malignancies exhibit increased radiosensitivity relative to other tumors due to impaired DNA repair mechanisms and heightened immunogenicity. Furthermore, understanding the interplay between HPV oncoproteins and p53 functionality can enhance treatment strategies for HPV-related cancers. Using DNA damage response inhibitors (PARP, ATM/ATR), cell cycle checkpoint inhibitors (WEE1, CHK1/2), and hypoxia-targeted agents as radiosensitizing strategies exhibit considerable promise. Immunomodulatory approaches, including PD-1 and CTLA-4 inhibitors in conjunction with radiation, enhance anti-tumor immunity. Future directions emphasize personalized radiation therapy using genetics, sophisticated medication delivery systems, adaptive radiotherapy, and real-time monitoring. These integrated strategies seek to diminish radiation resistance and improve therapeutic efficacy in HNSCC.
Collapse
Affiliation(s)
- Aastha Sobti
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Heath Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Christopher T Wilke
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA..
| |
Collapse
|
92
|
Altea-Manzano P, Decker-Farrell A, Janowitz T, Erez A. Metabolic interplays between the tumour and the host shape the tumour macroenvironment. Nat Rev Cancer 2025; 25:274-292. [PMID: 39833533 DOI: 10.1038/s41568-024-00786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Metabolic reprogramming of cancer cells and the tumour microenvironment are pivotal characteristics of cancers, and studying these processes offer insights and avenues for cancer diagnostics and therapeutics. Recent advancements have underscored the impact of host systemic features, termed macroenvironment, on facilitating cancer progression. During tumorigenesis, these inherent features of the host, such as germline genetics, immune profile and the metabolic status, influence how the body responds to cancer. In parallel, as cancer grows, it induces systemic effects beyond the primary tumour site and affects the macroenvironment, for example, through inflammation, the metabolic end-stage syndrome of cachexia, and metabolic dysregulation. Therefore, understanding the intricate metabolic interplay between the tumour and the host is a growing frontier in advancing cancer diagnosis and therapy. In this Review, we explore the specific contribution of the metabolic fitness of the host to cancer initiation, progression and response to therapy. We then delineate the complex metabolic crosstalk between the tumour, the microenvironment and the host, which promotes disease progression to metastasis and cachexia. The metabolic relationships among the host, cancer pathogenesis and the consequent responsive systemic manifestations during cancer progression provide new perspectives for mechanistic cancer therapy and improved management of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Ayelet Erez
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
93
|
Shiomura M, Watanabe T, Yasuda S, Fukuda I, Yamada T, Shichiri M. Refractory Hypoglycemia Secondary to the Warburg Effect in Diffuse Large B-Cell Lymphoma. JCEM CASE REPORTS 2025; 3:luaf038. [PMID: 40103858 PMCID: PMC11911912 DOI: 10.1210/jcemcr/luaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Indexed: 03/20/2025]
Abstract
Spontaneous and refractory hypoglycemia in malignancy poses diagnostic challenges, since its exact underlying mechanisms remain unclear. A 62-year-old female patient with a 10-year type 2 diabetes mellitus history presented with abdominal pain and spontaneous hypoglycemia despite discontinuation of her diabetic treatments. An initial computed tomography (CT) scan revealed a large perinephric tumor, and a second CT, performed a week later, demonstrated significant tumor growth. On admission, she had no neuroglycopenic symptoms despite a serum glucose level of 25 mg/dL (1.39 mmol/L). She showed suppressed insulin and insulin-like growth factor (IGF)-1 levels, elevated lactate levels, a pH of 7.434 with an anion gap of 24.1, and a negative test for anti-insulin antibody. A percutaneous CT-guided tumor biopsy revealed diffuse large B-cell lymphoma. She received continuous dextrose supplementation and prednisolone to alleviate the severe hypoglycemia, but she died from the tumor burden on the sixth day of hospitalization. Postmortem serum immunoblotting revealed the absence of partially processed IGF-2 precursors. The patient's refractory hypoglycemia and hyperlactatemia were consistent with tumor-associated aerobic glycolytic lactate production, known as the Warburg effect. This case illustrates the importance of increased awareness of this underrecognized oncologic emergency in the differential diagnosis of profound spontaneous hypoglycemia in malignancy.
Collapse
Affiliation(s)
- Miho Shiomura
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Kyosai Hospital, Tokyo 153-8934, Japan
| | - Takako Watanabe
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Kyosai Hospital, Tokyo 153-8934, Japan
| | - Shunichiro Yasuda
- Department of Hematology, Tokyo Kyosai Hospital, Tokyo 153-8934, Japan
| | - Izumi Fukuda
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Institute of Science Tokyo, Tokyo 113-8519, Japan
| | - Masayoshi Shichiri
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Kyosai Hospital, Tokyo 153-8934, Japan
| |
Collapse
|
94
|
Ren H, Tang Y, Zhang D. The emerging role of protein L-lactylation in metabolic regulation and cell signalling. Nat Metab 2025; 7:647-664. [PMID: 40175761 DOI: 10.1038/s42255-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025]
Abstract
L-Lactate has emerged as a crucial metabolic intermediate, moving beyond its traditional view as a mere waste product. The recent discovery of L-lactate-driven protein lactylation as a post-translational modification has unveiled a pathway that highlights the role of lactate in cellular signalling. In this Perspective, we explore the enzymatic and metabolic mechanisms underlying protein lactylation and its impacts on both histone and non-histone proteins in the contexts of physiology and diseases. We discuss growing evidence suggesting that this modification regulates a wide range of cellular functions and is involved in various physiological and pathological processes, such as cell-fate determination, development, cardiovascular diseases, cancer and autoimmune disorders. We propose that protein lactylation acts as a pivotal mechanism, integrating metabolic and signalling pathways to enable cellular adaptation, and highlight its potential as a therapeutic target in various diseases.
Collapse
Affiliation(s)
- Haowen Ren
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Yuwei Tang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Di Zhang
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
95
|
Zeng Q, Li L, Chang T, Sun Y, Zheng B, Xue L, Liu C, Li X, Huang R, Gu J, An Z, Yao H, Zhou D, Fan J, Dai Y. Phosphorylation of POU3F3 Mediated Nuclear Translocation Promotes Proliferation in Non-Small Cell Lung Cancer through Accelerating ATP5PF Transcription and ATP Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411503. [PMID: 39932442 PMCID: PMC11967767 DOI: 10.1002/advs.202411503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Indexed: 04/05/2025]
Abstract
Targeting oxidative phosphorylation (OXPHOS) through inhibiting the electron transport chain (ETC) has shown promising pre-clinical efficacy in cancer therapy. Although aerobic glycolysis is a hallmark of cancer, emerging evidence suggest OXPHOS is frequently enhanced, providing metabolic advantages for cell proliferation, metastasis, and drug resistance in a variety of aggressive cancer types including non-small cell lung cancer (NSCLC), yet the underlying molecular mechanisms remain elusive. Here it is reported that POU-domain containing family protein POU3F3 is translocated into the nuclei of NSCLC cell lines harboring mutant RAS, where it activates transcription of ATP5PF, an essential component of mitochondrial ATP synthase and consequent ATP production, leading to enhanced NSCLC proliferation and migration. Moreover, it is further found out that ERK1 phosphorylates POU3F3 at the S393 site in the cytoplasm and promotes the nuclear translocation of POU3F3 via receptor importin β1 in RAS mutant NSCLC cells. Mechanistically, RNA sequencing analysis combined with chromatin immunoprecipitation (ChIP) assay revealed that POU3F3 binds to the promoter of ATP5PF, leading to enhanced ATP5PF transcription and ATP production. Together, this study uncovers a novel RAS-POU3F3-ATP5PF axis in facilitating NSCLC progression, providing a new perspective on the understanding of molecular mechanisms for NSCLC progression.
Collapse
Affiliation(s)
- Qi‐Gang Zeng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Le Li
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Tao Chang
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Sun
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Bin Zheng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Ling‐Na Xue
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Chao‐Ling Liu
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Xia‐Qing Li
- Institute of Nephrology and Blood PurificationThe First Affiliated HospitalJinan UniversityGuangdong510632China
- Nephrology departmentThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityGuangdong517000China
| | - Ruo‐Tong Huang
- Department of Metabolism, Digestion, and ReproductionFaculty of MedicineImperial College LondonLondonW12 0NNUK
| | - Jia‐Xin Gu
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Zhao‐Rong An
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Hao‐Tao Yao
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Dan‐Yang Zhou
- Department of RespiratoryNanjing First HospitalNanjing Medical UniversityJiangsu210012China
| | - Jun Fan
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Dai
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| |
Collapse
|
96
|
Ocak M, Ateş Ş, Kahveci S, Okan A, Doğanyiğit Z, Uçar S, Yılmaz S. Evaluation of the anticarcinogenic effects of Rutin on brain tissue in mice with Ehrlich ascites carcinoma by micro-computed tomography and histological methods. Asia Pac J Clin Oncol 2025; 21:174-179. [PMID: 38526529 DOI: 10.1111/ajco.14058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Studies for new treatment strategies on cancer continue, and new searches continue in the diagnosis and evaluation of cancer. This study examined the possible anticarcinogenic effect of Rutin on the brain tissues of male mice with Ehrlich ascites carcinoma (EAC). MATERIAL AND METHODS We used micro-computed tomography (micro-CT) and histologically Hematoxylin&Eosin (H&E) staining methods for evaluation. RESULTS In the evaluation results, we saw a significant decrease in the brain volume of the tumor group to the control group. The difference in volume between the Rutin treatment group and the control group was not significant. In the brain tissues of the tumor group, numerous degenerated neurons characterized by pericellular/perivascular space expansion, cell swelling, or expansion were detected in the cortex and hippocampus regions. We showed a reduction in the damage rate in the Rutin treated group. CONCLUSION As a result, Rutin was found to have an anticarcinogenic effect. In addition to the classical histological evaluation, we used a newer method, micro-CT, in our study. We believe that this study has important results both in terms of its originality and adding new information to the literature.
Collapse
Affiliation(s)
- Mert Ocak
- Department of Anatomy, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Şükrü Ateş
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Selda Kahveci
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Aslı Okan
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Züleyha Doğanyiğit
- Department of Histology and Embriology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Sümeyye Uçar
- Department of Anatomy, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Seher Yılmaz
- Department of Anatomy, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
97
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
98
|
Asamitsu S, Ozawa Y, Okamoto H, Ogata S, Matsunaga T, Yoshitake J, Fusegawa K, Taniyama Y, Sato C, Ishida H, Abe T, Motohashi H, Akaike T, Kamei T. Supersulfide metabolome of exhaled breath condensate applied as diagnostic biomarkers for esophageal cancer. Cancer Sci 2025; 116:1023-1033. [PMID: 39895210 PMCID: PMC11967250 DOI: 10.1111/cas.16430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/11/2024] [Accepted: 11/29/2024] [Indexed: 02/04/2025] Open
Abstract
Early detection of esophageal cancer is essential for esophagogastroduodenoscopy and histopathological diagnosis. However, endoscopic examinations are sometimes invasive, which limits their clinical application and compliance, and traditional blood tumor markers are unsuitable for cancer screening. The current study aimed to evaluate the usefulness of sulfur metabolites as new biomarkers for esophageal cancer using blood samples and exhaled breath condensate (EBC), which can be readily obtained and is non-invasive. We collected EBC and plasma samples from 50 patients with esophageal cancer and 30 healthy controls. Sulfur metabolome analysis using tandem mass spectrometry was performed to compare the metabolic profile between the two groups. Supersulfide metabolic profiles were different between the two cohorts. Supersulfide metabolome analysis showed that cysteine hydropersulfide (CysSSH) and homocysteine hydropersulfide (HomoCysSSH) were increased in the plasma of patients with esophageal cancer. Elevated levels of HomoCysSSH could distinguish patients with esophageal cancer from healthy subjects (area under the curve [AUC]: 0.93, sensitivity: 89%, specificity: 96%). Interestingly, we also detected an elevation of supersulfides in the EBC analysis. CysSSH levels significantly increased in the EBC recovered from patients with esophageal cancer (AUC: 0.71, sensitivity: 60%, specificity: 96%). In addition, the observed level was correlated with that of HomoCysSSH in the plasma (r = 0.27). Supersulfides, such as CysSSH and HomoCysSSH, are potential biomarkers for detecting esophageal cancer. CysSSH from EBC may serve as a valuable non-invasive biomarker with similar detection ability but with superior precision and convenience compared with the currently available blood biomarkers.
Collapse
Affiliation(s)
- Seji Asamitsu
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Yohei Ozawa
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroshi Okamoto
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Seiryo Ogata
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Jun Yoshitake
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kazuki Fusegawa
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Yusuke Taniyama
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Chiaki Sato
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Hirotaka Ishida
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal RegulationTohoku University Graduate School of MedicineSendaiJapan
| | - Hozumi Motohashi
- Department of Medical BiochemistryTohoku University Graduate School of MedicineSendaiJapan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Takashi Kamei
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
99
|
Huang Y, Zhu H, Liang Z, Wei W, Yang H, Wang Q, Huang H, He H, Mo R, Ye J, Dai Q, Zhong W, Liang Y. Development and validation of a kinase-related gene signature as a novel diagnostic and prognostic model for prostate cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167722. [PMID: 39965532 DOI: 10.1016/j.bbadis.2025.167722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Prostate cancer (PCa) is a prevalent malignant tumor in men worldwide. Kinases play a key role in the development of multiple tumors. Nevertheless, the role of kinases in PCa remains largely unclear. METHODS A kinase-related gene signature was constructed by LASSO Cox regression analysis using the TCGA_PRAD cohort. The diagnostic and prognostic values of the signature were then evaulated. Furthermore, a loss-of-function assay was carried out to explore the function of NEK5 in PCa. RESULTS A signature of 13 kinase-related genes (NEK5, FRK, STK39, STYK1, IGF1R, RPS6KC1, TTK, CDK1, NEK2, PTK6, DAPK1, MELK and EPHA10) was constructed. The PCa patients presenting a high-risk score according to the signature demonstrated poorer disease-free survival compared to those with a low score. Additionally, TMB was found to be remarkably increased in patients categorized as high-risk relative to low-risk patients. Moreover, the 13-gene signature may also have good predictive value for PCa diagnosis. Furthermore, NEK5 expression was remarkably elevated in PCa tissues relative to benign tissues. NEK5 deficiency significantly inhibited PCa cell growth and suppressed mitochondrial OXPHOS. CONCLUSION The 13-gene signature constructed in this study may exhibit good performance in PCa diagnosis and prognosis evaluation. We identified the oncogenic role of NEK5 in PCa. NEK5 may serve as a therapeutic target for treatting PCa.
Collapse
Affiliation(s)
- Yaqiang Huang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Haiying Zhu
- Department of Endocrinology and Metabolism, The Affiliated Shunde Hospital of Jinan University, Foshan 528305, Guangdong, China
| | - Zhenguo Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| | - Weiyang Wei
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Hao Yang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Qi Wang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Hongxing Huang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Huichan He
- Guangzhou national laboratory, Guangzhou 510120, Guangdong, China
| | - Rujun Mo
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan People's Hospital, Dongguan 523018, Guangdong, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China
| | - Qishan Dai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China
| | - Weide Zhong
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China; Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau.
| | - Yingke Liang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
100
|
Jalil AT, Al-Kazzaz HH, Hassan FA, Mohammed SH, Merza MS, Aslandook T, Elewadi A, Fadhil A, Alsalamy A. Metabolic Reprogramming of Anti-cancer T Cells: Targeting AMPK and PPAR to Optimize Cancer Immunotherapy. Indian J Clin Biochem 2025; 40:165-175. [PMID: 40123631 PMCID: PMC11928344 DOI: 10.1007/s12291-023-01166-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/17/2023] [Indexed: 03/25/2025]
Abstract
Cancer treatment era has been revolutionized by the novel therapeutic methods such as immunotherapy in recent years. Immunotherapy-based approaches are considered effective and reliable methods that has brought hope to eradicate certain cancers. Nonetheless, there are some issues, considered as critical obstacles in successful cancer immunotherapy. Such issues are attributed to the ability of the tumor cells in providing a tolerant microenvironment that impairs the immune responses, and help the cancer cells evade the immunogenic cell death. It has been suggested that the re-activation and maintenance of effector immune cells may become possible by metabolic reprogramming. Several signaling pathways have been noticed with the possibility of metabolic reprogramming of tumor-specific T cells, to overcome the metabolic restrictions in the tumor microenvironment; and among them, AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptors (PPAR) have been investigated the most as the main energy sensors and regulators of mitochondrial biogenesis. The synergic effects of AMPK activators and/or PPAR agonists in cancer immunotherapy have been reported. In this review, we compare the roles of AMPK activators and PPAR agonists, and the efficacy of their combination in metabolic reprogramming of cytotoxic T cells in favoring cancer immunotherapy.
Collapse
Affiliation(s)
| | - Hassan Hadi Al-Kazzaz
- College of Medical and Health Technology, Al-Zahraa University for Women, Karbala, Iraq
| | - Firas A. Hassan
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | | | - Muna S. Merza
- Department of Prosthetic Dental Techniques, Al-Mustaqbal University College, Hillah, Iraq
| | - Tahani Aslandook
- Department of Dentistry, Al-Turath University College, Baghdad, Iraq
| | - Ahmed Elewadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Ali Fadhil
- College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja’afar Al-Sadiq University, Al-Muthanna, 66002 Iraq
| |
Collapse
|