101
|
Dong J, Konopleva M. Preclinical targeting of leukemia-initiating cells in the development future biologics for acute myeloid leukemia. Expert Opin Ther Targets 2025; 29:223-237. [PMID: 40304258 DOI: 10.1080/14728222.2025.2500417] [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: 11/16/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Leukemia-initiating cells (LICs) are a critical subset of cells driving acute myeloid leukemia (AML) relapse and resistance to therapy. They possess unique properties, including metabolic, epigenetic, and microenvironmental dependencies, making them promising therapeutic targets. AREAS COVERED This review summarizes preclinical advances in targeting AML LICs, including strategies to exploit metabolic vulnerabilities, such as the reliance on oxidative phosphorylation (OXPHOS), through the use of mitochondrial inhibitors; target epigenetic regulators like DOT1L (Disruptor of Telomeric Silencing 1-like) to disrupt LIC survival mechanisms; develop immunotherapies, including CAR (chimeric antigen receptor) T-cell therapy, and bispecific antibodies; and disrupt LIC interactions with the bone marrow microenvironment by inhibiting supportive niches. EXPERT OPINION LIC-targeted therapies hold significant promise for revolutionizing AML treatment by reducing relapse rates and improving long-term outcomes. However, challenges such as LIC heterogeneity, therapy resistance, and associated toxicity persist. Recent studies have illuminated the distinct biological characteristics of LICs, advancing our understanding of their behavior and vulnerabilities. These insights offer new opportunities to target LICs at earlier disease stages and to explore combination therapies with other targeted treatments, ultimately enhancing therapeutic efficacy and improving patient outcomes.
Collapse
Affiliation(s)
- Jiaxin Dong
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
102
|
Lee YJ, Seo CW, Chae S, Lee CY, Kim SS, Shin YH, Park HM, Gho YS, Ryu S, Lee SH, Choi D. Metabolic Reprogramming Into a Glycolysis Phenotype Induced by Extracellular Vesicles Derived From Prostate Cancer Cells. Mol Cell Proteomics 2025; 24:100944. [PMID: 40089067 PMCID: PMC12008616 DOI: 10.1016/j.mcpro.2025.100944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Most cancer cells adopt a less efficient metabolic process of aerobic glycolysis with high level of glucose uptake followed by lactic acid production, known as the Warburg effect. This phenotypic transition enables cancer cells to achieve increased cellular survival and proliferation in a harsh low-oxygen tumor microenvironment. Also, the resulting acidic microenvironment causes inactivation of the immune system such as T-cell impairment that favors escape by immune surveillance. While lots of studies have revealed that tumor-derived EVs can deliver parental materials to adjacent cells and contribute to oncogenic reprogramming, their functionality in energy metabolism is not well addressed. In this study, we established prostate cancer cells PC-3AcT resistant to cellular death in an acidic culture medium driven by lactic acid. Quantitative proteomics between EVs derived from PC-3 and PC-3AcT cells identified 935 confident EV proteins. According to cellular adaptation to lactic acidosis, we revealed 159 regulated EV proteins related to energy metabolism, cellular shape, and extracellular matrix. These EVs contained a high abundance of glycolytic enzymes. In particular, PC-3AcT EVs were enriched with apolipoproteins including apolipoprotein B-100 (APOB). APOB on PC-3AcT EVs could facilitate their endocytic uptake depending on low density lipoprotein receptor of recipient PC-3 cells, encouraging increases of cellular proliferation and survival in acidic culture media via increased activity and expression of hexokinases and phosphofructokinase. The activation of recipient PC-3 cells can increase glucose consumption and ATP generation, representing an acquired metabolic reprogramming into the Warburg phenotype. Our study first revealed that EVs derived from prostate cancer cells could contribute to energy metabolic reprogramming and that the acquired metabolic phenotypic transition of recipient cells could favor cellular survival in tumor microenvironment.
Collapse
Affiliation(s)
- Yoon-Jin Lee
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea
| | - Chul Won Seo
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea
| | - Shinwon Chae
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea
| | - Chang Yeol Lee
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea
| | - Sang Soo Kim
- Department of Life Sciences, POSTECH, Pohang, Republic of Korea
| | - Yoon-Hee Shin
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, POSTECH, Pohang, Republic of Korea
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Sang-Han Lee
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea
| | - Dongsic Choi
- Department of Biochemistry, Soonchunhyang University, College of Medicine, Cheonan, Republic of Korea.
| |
Collapse
|
103
|
Zhang R, Jin W, Wang K. Glycolysis-Driven Prognostic Model for Acute Myeloid Leukemia: Insights into the Immune Landscape and Drug Sensitivity. Biomedicines 2025; 13:834. [PMID: 40299448 PMCID: PMC12024913 DOI: 10.3390/biomedicines13040834] [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: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Acute myeloid leukemia (AML), a malignant blood disease, is caused by the excessive growth of undifferentiated myeloid cells, which disrupt normal hematopoiesis and may invade several organs. Given the high heterogeneity in prognosis, identifying stable prognostic biomarkers is crucial for improved risk stratification and personalized treatment strategies. Although glycolysis has been extensively studied in cancer, its prognostic significance in AML remains unclear. Methods: Glycolysis-related prognostic genes were identified by differential expression profiles. We modeled prognostic risk by least absolute shrinkage and selection operator (LASSO) regression and validated it by Kaplan-Meier (KM) survival analysis, receiver operating characteristic (ROC) curves, and independent datasets (BeatAML2.0, GSE37642, GSE71014). Mechanisms were further explored through immune microenvironment analysis and drug sensitivity scores. Results: Differential expression and survival correlation analysis across the genes associated with glycolysis revealed multiple glycolytic genes associated with the outcomes of AML. We constructed a seven-gene prognostic model (G6PD, TFF3, GALM, SOD1, NT5E, CTH, FUT8). Kaplan-Meier analysis demonstrated significantly reduced survival in high-risk patients (hazard ratio (HR) = 3.4, p < 0.01). The model predicted the 1-, 3-, and 5-year survival outcomes, achieving area under the curve (AUC) values greater than 0.8. Immune profiling indicated distinct cellular compositions between risk groups: high-risk patients exhibited elevated monocytes and neutrophils but reduced Th1 cell infiltration. Drug sensitivity analysis showed that high-risk patients exhibited resistance to crizotinib and lapatinib but were more sensitive to motesanib. Conclusions: We established a novel glycolysis-related gene signature for AML prognosis, enabling effective risk classification. Combined with immune microenvironment analysis and drug sensitivity analysis, we screened metabolic characteristics and identified an immune signature to provide deeper insight into AML. Our findings may assist in identifying new therapeutic targets and more effective personalized treatment regimes.
Collapse
Affiliation(s)
- Rongsheng Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Wen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China; (R.Z.); (W.J.)
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd., Shanghai 200025, China
| |
Collapse
|
104
|
Chang MC, Malut VR, Mahar R, Rushin A, McLeod MA, Pierre GL, Malut IR, Staklinski SJ, Glanz ME, Ragavan M, Sharma G, Madheswaran M, Badar A, Rao AD, Law BK, Kilberg MS, Collins JHP, Kodibagkar VD, Bankson JA, DeBerardinis RJ, Merritt ME. Assessing cancer therapeutic efficacy in vivo using [ 2H 7]glucose deuterium metabolic imaging. SCIENCE ADVANCES 2025; 11:eadr0568. [PMID: 40138413 PMCID: PMC11939044 DOI: 10.1126/sciadv.adr0568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Metabolic imaging produces powerful visual assessments of organ function in vivo. Current techniques can be improved by safely increasing metabolic contrast. The gold standard, 2-[18F]fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging, is limited by radioactive exposure and sparse assessment of metabolism beyond glucose uptake and retention. Deuterium magnetic resonance imaging (DMRI) with [6,6-2H2]glucose is nonradioactive, achieves tumor metabolic contrast, but can be improved by enriched contrast from deuterated water (HDO) based imaging. Here, we developed a DMRI protocol employing [2H7]glucose. Imaging 2H-signal and measuring HDO production in tumor-bearing mice detected differential glucose utilization across baseline tumors, tumors treated with vehicle control or anti-glycolytic BRAFi and MEKi therapy, and contralateral healthy tissue. Control tumors generated the most 2H-signal and HDO. To our knowledge this is the first application of DMRI with [2H7]glucose for tumoral treatment monitoring. This approach demonstrates HDO as a marker of tumor glucose utilization and suggests translational capability in humans due to its safety, noninvasiveness, and suitability for serial monitoring.
Collapse
Affiliation(s)
- Mario C. Chang
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Vinay R. Malut
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Rohit Mahar
- Department of Chemistry, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand 246174, India
| | - Anna Rushin
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Marc A. McLeod
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Geraldine L. Pierre
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Indu R. Malut
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Stephen J. Staklinski
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Max E. Glanz
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mukundan Ragavan
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Gaurav Sharma
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Manoj Madheswaran
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Arshee Badar
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Aparna D. Rao
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Michael S. Kilberg
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - James H. P. Collins
- National High Magnetic Field Laboratory, University of Florida, Gainesville, FL 32610, USA
| | - Vikram D. Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA
| | - James A. Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ralph J. DeBerardinis
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
105
|
Wu T, Wu X. Construction and evaluation of a prognostic model based on the expression of the metabolism-related signatures in patients with osteosarcoma. BMC Musculoskelet Disord 2025; 26:303. [PMID: 40148931 PMCID: PMC11948978 DOI: 10.1186/s12891-025-08439-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The aim of this study was to screen three major substance metabolism-related genes and establish a prognostic model for osteosarcoma. METHODS RNA-seq expression data for osteosarcoma were downloaded from The Cancer Genome Atlas (TCGA) and GEO databases. Differentially expressed (DE) RNAs were selected, followed by the selection of metabolic-related DE mRNAs. Using Cox regression analysis, prognostic DE RNAs were identified to construct a prognostic model. Subsequently, independent prognostic clinical factors were screened, and the functions of the long non-coding RNAs (lncRNAs) were analyzed. Finally, the expression of signature genes was further tested in osteosarcoma cells using quantitative reverse transcription quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. RESULTS A total of 432 DE RNAs, comprising 79 DE lncRNAs and 353 DE mRNAs were obtained, and then 107 metabolic-related DE mRNAs. Afterwards signature genes (LINC00545, LINC01537, FOXC2-AS1, CYP27B1, PFKFB4, PHKG1, PHYKPL, PXMP2, and XYLB) served as optimal combinations, and a prognostic score model was successfully proposed. Three verification datasets (GSE16091, GSE21257, and GSE39055) showed that the model had high specificity and sensitivity. In addition, two independent prognostic clinical factors (age and tumor metastasis) were identified. Finally, the concordance rate between the in silico analysis, qRT-PCR, and western blotting analysis was 88.89% (8/9), suggesting the robustness of our analysis. CONCLUSIONS The prognostic model based on the nine signature genes accurately predicted the prognosis of patients with osteosarcoma; CYP27B1, PFKFB4, PHKG1, PHYKPL, PXMP2, and XYLB may serve as metabolism-related biomarkers in osteosarcoma.
Collapse
Affiliation(s)
- Tieli Wu
- Hainan Vocational University of Science and Technology, Hainan Province, Haikou, 570000, China
| | - Xingyi Wu
- Department of Internal Medicine, Qiqihar First Factory Hospital, 27 Xinming Street, Qiqihar, 161000, Heilongjiang Province, China.
| |
Collapse
|
106
|
Xi JF, Liu BD, Tang GR, Ren ZH, Chen HX, Lan YL, Yin F, Li Z, Cheng WS, Wang J, Chen L, Yuan SC, Zhang Z, Luo GZ. m 6A modification regulates cell proliferation via reprogramming the balance between glycolysis and pentose phosphate pathway. Commun Biol 2025; 8:496. [PMID: 40140553 PMCID: PMC11947274 DOI: 10.1038/s42003-025-07937-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
N6-methyladenosine (m6A) stands as the predominant modification in eukaryotic mRNA and is involved in various biological functions. Aberrant m6A has been implicated in abnormal cellular phenotypes, including defects in stem cell differentiation and tumorigenesis. However, the precise effects of m6A on cell proliferation and the underlining mechanism of metabolic gene regulation remain incompletely understood. Here, we established a cellular environment with low-m6A levels and observed a severe impairment of cell proliferation. Mechanistic studies revealed that the depletion of m6A on TIGAR mRNA led to increased expression, subsequently inhibiting glycolysis while promoting the pentose phosphate pathway (PPP). A genome-wide CRISPR-Cas9 screen identified numerous genes involved in cell proliferation that are sensitive to m6A modification, with G6PD emerging as a key regulator. Integration of gene expression and survival data from cancer patients suggested that patients with elevated G6PD expression may exhibit enhanced responsiveness to tumor growth inhibition through m6A suppression. Our findings elucidate the critical role of m6A in cell proliferation, highlighting the therapeutic potential of targeting m6A-mediated metabolic pathways in cancer.
Collapse
Affiliation(s)
- Jian-Fei Xi
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Biao-Di Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Guo-Run Tang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ze-Hui Ren
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hong-Xuan Chen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ye-Lin Lan
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Feng Yin
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, China
| | - Zigang Li
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, China
| | - Wei-Sheng Cheng
- Department of Medical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinkai Wang
- Department of Medical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lili Chen
- Guangdong Provincial Key Laboratory of Stomatology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shao-Chun Yuan
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhang Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Guan-Zheng Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
- Pingshan Translational Medicine Center, Shenzhen Bay Laboratory, Shenzhen, China.
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, China.
| |
Collapse
|
107
|
Martinez P, Sabatier JM. Rethinking corticosteroids use in oncology. Front Pharmacol 2025; 16:1551111. [PMID: 40206059 PMCID: PMC11979161 DOI: 10.3389/fphar.2025.1551111] [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: 12/24/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Corticosteroids (CSs), widely used in oncology for their anti-inflammatory and immunosuppressive properties, help manage cancer-related symptoms and side effects. However, their long-term use may negatively affect patient survival and exacerbate tumor progression. Elevated glucose and glutamine metabolism, disruption of vitamin D levels, and alterations in the microbiome are some of the key factors contributing to these adverse outcomes. Approaches such as ketogenic diets, fasting, sartans, and vitamin D supplementation have shown promise in providing similar benefits to CSs while mitigating the risks associated with the mechanisms identified as contributing to tumor progression. This perspective underscores the necessity for a reevaluation of CSs use in cancer care and advocates for further research into safer, more effective therapeutic strategies.
Collapse
Affiliation(s)
| | - Jean-Marc Sabatier
- Institut de NeuroPhysiopathologie (INP), CNRS UMR 7051, Marseille, France
| |
Collapse
|
108
|
Kanamori T, Yasuda S, Duan R, Ohashi M, Amou M, Hori K, Tsuda R, Fujimoto T, Higashi K, Xu W, Niidome T, Hatakeyama H. Cholesterol depletion suppresses thermal necrosis resistance by alleviating an increase in membrane fluidity. Sci Rep 2025; 15:10133. [PMID: 40128234 PMCID: PMC11933367 DOI: 10.1038/s41598-025-92232-0] [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: 11/20/2024] [Accepted: 02/26/2025] [Indexed: 03/26/2025] Open
Abstract
Thermally resistant cancer cells suppress the therapeutic effects of hyperthermia. However, the mechanism underlying the thermal resistance remains unclear. With the aim of enhancing the therapeutic effects of hyperthermia, we investigated the mechanism underlying thermal resistance. We found that heat shock-induced cell death can be classified into two types: late-phase apoptosis and early-phase necrosis. Cell death was suppressed in thermally resistant cells. In addition, heat-induced necrosis resistance correlated with plasma membrane fluidity, which was maintained by cholesterol. Depletion of cholesterol from cancer cells and tumor tissues enhanced the effect of hyperthermia under both in vivo and in vitro conditions. Hence, the findings demonstrate the usefulness of cholesterol as a marker for thermally resistant cancer cells. Furthermore, the combination of cholesterol depletion and hyperthermia may be a new therapeutic strategy for thermally resistant cancers.
Collapse
Affiliation(s)
- Taisei Kanamori
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Shogo Yasuda
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Runjing Duan
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Mei Ohashi
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Mai Amou
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Kanato Hori
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Ryota Tsuda
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Taiki Fujimoto
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Kenjirou Higashi
- Laboratory of Pharmaceutical Technology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan
| | - Wei Xu
- Faculty of Advanced Science and Technology, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Graduate School of Science and Technology, Kumamoto University, Kumamoto, 860-8555, Japan
| | - Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-0856, Japan.
| |
Collapse
|
109
|
Gautam V, Saini A, Misra A, Trivedi NK, Maheshwari S, Gaurang Tiwari R. Parallel convolutional SpinalNet: A hybrid deep learning approach for breast cancer detection using mammogram images. NETWORK (BRISTOL, ENGLAND) 2025:1-41. [PMID: 40125951 DOI: 10.1080/0954898x.2025.2480299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Breast cancer is the foremost cause of mortality among females. Early diagnosis of a disease is necessary to avoid breast cancer by reducing the death rate and offering a better life to the individuals. Therefore, this work proposes a Parallel Convolutional SpinalNet (PConv-SpinalNet) for the efficient detection of breast cancer using mammogram images. At first, the input image is pre-processed using the Gabor filter. The tumour segmentation is conducted using LadderNet. Then, the segmented tumour samples are augmented using Image manipulation, Image erasing, and Image mix techniques. After that, the essential features, like CNN features, Texton, Local Gabor binary patterns (LGBP), scale-invariant feature transform (SIFT), and Local Monotonic Pattern (LMP) with discrete cosine transform (DCT) are extracted in the feature extraction phase. Finally, the detection of breast cancer is performed using PConv-SpinalNet. PConv-SpinalNet is developed by an integration of Parallel Convolutional Neural Networks (PCNN) and SpinalNet. The evaluation results show that PConv-SpinalNet accomplished a superior range of accuracy as 88.5%, True Positive Rate (TPR) as 89.7%, True Negative Rate (TNR) as 90.7%, Positive Predictive Value (PPV) as 91.3%, and Negative Predictive Value (NPV) as 92.5%.
Collapse
Affiliation(s)
- Vinay Gautam
- Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Anu Saini
- Department of Computer Science and Engineering, G.B. Pant DSEU OKHLA-I Campus, New Delhi, India
| | - Alok Misra
- School of Computer Science and Engineering, Lovely Professional University, Phagwara, India
| | - Naresh Kumar Trivedi
- Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| | - Shikha Maheshwari
- Centre for Distance & Online Education, Manipal University, Jaipur, India
| | - Raj Gaurang Tiwari
- Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
| |
Collapse
|
110
|
Stea DM, D’Alessio A. Caveolae: Metabolic Platforms at the Crossroads of Health and Disease. Int J Mol Sci 2025; 26:2918. [PMID: 40243482 PMCID: PMC11988808 DOI: 10.3390/ijms26072918] [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: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dante Maria Stea
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Alessio D’Alessio
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy
| |
Collapse
|
111
|
Hanusrichterova J, Baranovicova E, Barosova R, Kolomaznik M, Mikolka P, Kosutova P, Mokra D, Mokry J, Calkovska A. Metabolic profiling in experimental guinea pig models of bacterial and allergic inflammation. Metabolomics 2025; 21:43. [PMID: 40123009 PMCID: PMC11930882 DOI: 10.1007/s11306-025-02239-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Based on distinct triggers, bacterial and allergen-induced inflammatory reactions have different pathophysiology. Metabolomic analysis is high-throughput technique that can provide potential biomarkers to distinguish between these responses. OBJECTIVES In order to find out the metabolic profiles of two types of inflammation, metabolites were analysed in blood plasma and bronchoalveolar lavage fluid (BALF) of guinea pigs subjected to bacterial lipopolysaccharide (LPS) or allergen ovalbumin (OVA). METHODS Hydrogen-1 nuclear magnetic resonance (1H NMR) spectroscopy for metabolite analysis was performed in samples of blood plasma and BALF of guinea pigs. RESULTS Random forest algorithm built on combination of levels of circulating and BALF metabolites resulted in almost ideal discrimination between acute allergic and bacterial inflammation. The differences between inflammation triggered by LPS and OVA were manifested in shift in energy metabolism, metabolism of branched-chain amino acids (BCAAs)/branched-chain keto acids (BCKAs) with alterations in alanine and glutamine, which are linked with both, ammonia homeostasis as well as gluconeogenesis. CONCLUSION Distinct molecule nutrients are to be utilized during acute bacterial and allergic inflammatory response.
Collapse
Affiliation(s)
- J Hanusrichterova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4D, 03601, Martin, Slovakia.
| | - E Baranovicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4D, 03601, Martin, Slovakia
| | - R Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4C, 03601, Martin, Slovakia
| | - M Kolomaznik
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4D, 03601, Martin, Slovakia
| | - P Mikolka
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4C, 03601, Martin, Slovakia
| | - P Kosutova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4D, 03601, Martin, Slovakia
| | - D Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4C, 03601, Martin, Slovakia
| | - J Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4C, 03601, Martin, Slovakia
| | - A Calkovska
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 11161/4C, 03601, Martin, Slovakia
| |
Collapse
|
112
|
Baldassari F, Bonanomi M, Mallia S, Bonas M, Brivio E, Aramini T, Porro D, Gaglio D. Emodin and Aloe-Emodin Reduce Cell Growth and Disrupt Metabolic Plasticity in Human Melanoma Cells. Nutrients 2025; 17:1113. [PMID: 40218871 PMCID: PMC11990439 DOI: 10.3390/nu17071113] [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: 01/13/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Melanoma is an aggressive skin cancer with intratumor metabolic heterogeneity, which drives its progression and therapy resistance. Natural anthraquinones, such as emodin and aloe-emodin, exhibit anti-cancer properties, but their effects on metabolic plasticity remain unclear. This study evaluated their impact on proliferation and metabolic pathways in heterogenous melanoma human cell lines. Methods: COLO 800, COLO 794, and A375 melanoma cell lines representing distinct metabolic phenotypes were analyzed. Targeted and untargeted metabolomics analyses integrated with Seahorse assays were performed to assess the effects of emodin and aloe-emodin on cell proliferation, mitochondrial function, and redox homeostasis. Glucose tracing using [U-13C6] glucose and metabolic flux analysis (MFA) were carried out to evaluate the glycolysis and TCA cycle dynamics. Results: Emodin and aloe-emodin inhibited proliferation by disrupting glycolysis, oxidative phosphorylation, and energy production across all cell lines. Both compounds impaired glucose metabolism, reduced TCA cycle intermediates, and induced mitochondrial ROS accumulation, causing oxidative stress and redox imbalance. Despite intrinsic metabolic differences, COLO 800 and COLO 794 upregulated antioxidant defenses; A375 enhanced one-carbon metabolism and amino acid pathways to maintain redox balance and nucleotide biosynthesis. Conclusions: Emodin and aloe-emodin can disrupt the metabolic plasticity of melanoma cells by impairing glycolysis, mitochondrial function, and redox homeostasis. Their ability to target metabolic vulnerabilities across diverse phenotypes highlights their therapeutic potential for overcoming resistance mechanisms and advancing melanoma treatment strategies.
Collapse
Affiliation(s)
- Federica Baldassari
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, PA, Italy
| | - Marcella Bonanomi
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
| | - Sara Mallia
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, PA, Italy
| | - Matteo Bonas
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, MI, Italy; (M.B.); (E.B.)
| | - Elisa Brivio
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, MI, Italy; (M.B.); (E.B.)
| | - Tecla Aramini
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
| | - Danilo Porro
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, PA, Italy
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, 20126 Milano, MI, Italy; (M.B.); (E.B.)
| | - Daniela Gaglio
- Institute of Bioimaging and Complex Biological Systems, National Research Council (CNR), 20054 Segrate, MI, Italy; (F.B.); (M.B.); (S.M.); (T.A.); (D.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, PA, Italy
| |
Collapse
|
113
|
Ma J, Tang L, Xiao J, Tang K, Zhang H, Huang B. Burning lactic acid: a road to revitalizing antitumor immunity. Front Med 2025:10.1007/s11684-025-1126-6. [PMID: 40119026 DOI: 10.1007/s11684-025-1126-6] [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: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/24/2025]
Abstract
Lactic acid (LA) accumulation in tumor microenvironments (TME) has been implicated in immune suppression and tumor progress. Diverse roles of LA have been elucidated, including microenvironmental pH regulation, signal transduction, post-translational modification, and metabolic remodeling. This review summarizes LA functions within TME, focusing on the effects on tumor cells, immune cells, and stromal cells. Reducing LA levels is a potential strategy to attack cancer, which inevitably affects the physiological functions of normal tissues. Alternatively, transporting LA into the mitochondria as an energy source for immune cells is intriguing. We underscore the significance of LA in both tumor biology and immunology, proposing the burning of LA as a potential therapeutic approach to enhance antitumor immune responses.
Collapse
Affiliation(s)
- Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| | - Liang Tang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jingxuan Xiao
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Ke Tang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Huang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| |
Collapse
|
114
|
Onyiba CI, Kumar NK, Scarlett CJ, Weidenhofer J. Cell Progression and Survival Functions of Enzymes Secreted in Extracellular Vesicles Associated with Breast and Prostate Cancers. Cells 2025; 14:468. [PMID: 40214422 PMCID: PMC11988166 DOI: 10.3390/cells14070468] [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] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound cargoes secreted by normal and pathological cells. Through their protein, nucleic acid, and lipid cargoes, EVs mediate several cellular processes, such as cell-cell communication, cell development, immune response, and tissue repair. Most importantly, through their enzyme cargo, EVs mediate pathophysiological processes, including the pathogenesis of cancer. In this review, we enumerate several enzymes secreted in EVs (EV enzyme cargo) from cells and patient clinical samples of breast and prostate cancers and detail their contributions to the progression and survival of both cancers. Findings in this review reveal that the EV enzyme cargo could exert cell progression functions via adhesion, proliferation, migration, invasion, and metastasis. The EV enzyme cargo might also influence cell survival functions of chemoresistance, radioresistance, angiogenesis, cell death inhibition, cell colony formation, and immune evasion. While the current literature provides evidence of the possible contributions of the EV enzyme cargo to the progression and survival mechanisms of breast and prostate cancers, future studies are required to validate that these effects are modified by EVs and provide insights into the clinical applications of the EV enzyme cargo in breast and prostate cancer.
Collapse
Affiliation(s)
- Cosmos Ifeanyi Onyiba
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Niwasini Krishna Kumar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Christopher J. Scarlett
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Ourimbah, NSW 2258, Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
115
|
Yi D, Zhou K, Pan Y, Cai H, Huang P. The lactylation modification of proteins plays a critical role in tumor progression. Front Oncol 2025; 15:1530567. [PMID: 40190564 PMCID: PMC11970033 DOI: 10.3389/fonc.2025.1530567] [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: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Lactylation modifications have been shown to be a novel type of protein post-translational modifications (PTMs), providing a new perspective for understanding the interaction between cellular metabolic reprogramming and epigenetic regulation. Studies have shown that lactylation plays an important role in the occurrence, development, angiogenesis, invasion and metastasis of tumors. It can not only regulate the phenotypic expression and functional polarization of immune cells, but also participate in the formation of tumor drug resistance through a variety of molecular mechanisms. In this review, we review the latest research progress of lactylation modification in tumors, focusing on its mechanism of action in angiogenesis, immune cell regulation in tumor microenvironment (TME), and tumor drug resistance, aiming to provide a theoretical basis and research ideas for the discovery of new therapeutic targets and methods. Through the in-depth analysis of lactylation modification, it is expected to open up a new research direction for tumor treatment and provide potential strategies for overcoming tumor drug resistance and improving clinical efficacy.
Collapse
Affiliation(s)
- Dehao Yi
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yinlong Pan
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Pan Huang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
116
|
Kumari S, Makarewicz A, Klubo-Gwiezdzinska J. Emerging Potential of Metabolomics in Thyroid Cancer-A Comprehensive Review. Cancers (Basel) 2025; 17:1017. [PMID: 40149351 PMCID: PMC11940765 DOI: 10.3390/cancers17061017] [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: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Thyroid cancer is a very common endocrine system malignancy. Nevertheless, a dearth of precise markers makes it challenging to apply precision medicine to thyroid cancer. The limitations of standard diagnosis techniques (fine-needle aspiration biopsy), such as indeterminate cases and inaccuracies in distinguishing between different types of cancers, lead to unnecessary surgeries and thus warrant the development of more discriminatory biomarkers to improve the accuracy of existing diagnostic and prognostic techniques. Moreover, individualized therapies for thyroid cancer are necessary to avoid overtreatment of indolent lesions and undertreatment of high-risk progressive disease. As thyroid cancer metabolic signatures are associated with disease aggressiveness and responsiveness to therapy, metabolomics has been recently used for diagnostic and prognostic biomarker discovery. This strategy has enabled the detection of several metabolites from tissue samples or biofluids to facilitate the classification of disease aggressiveness and to potentially assist in individualized therapies. In this review, we summarize the utilization and potential of metabolomics in thyroid cancer.
Collapse
Affiliation(s)
| | | | - Joanna Klubo-Gwiezdzinska
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (S.K.); (A.M.)
| |
Collapse
|
117
|
Ge H, Yan S, Yin M, Gao Y, Wang J, Wang Q, Xu G, Yang M. Gua Sha Alleviates Radiculitis-Induced Pain Via HIF-1α-Mediated Metabolic Reprogramming Pathway in Rats. Pain Res Manag 2025; 2025:9923147. [PMID: 40130025 PMCID: PMC11932754 DOI: 10.1155/prm/9923147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Background: Radiculitis-induced pain (RIP) results from dorsal root ganglion (DRG) sensitization due to inflammation. Hypoxia-inducible factor 1-alpha (HIF-1α) is linked to inflammatory responses through metabolic changes, but its role in RIP is not well understood. Gua Sha therapy has been shown to reduce inflammation and neural damage from lumbar disc herniation (LDH). This study investigates whether HIF-1α-mediated metabolic reprogramming contributes to the pain-relieving effects of Gua Sha in RIP. Methods: Male SD rats were subjected to LDH surgery and divided into six groups: sham, model, sham Gua Sha, Gua Sha, Gua Sha + DMOG, and Gua Sha + YC-1. Gua Sha was applied 5 days postsurgery, every other day for three sessions per course, totaling three courses. Changes in paw withdrawal threshold (PWT) and latency (PWL) were monitored, along with blood flow in the rats' backs. Levels of IL-1β, TNF-α, and NF-κB were assessed in serum and DRG tissue. Pathological changes and hypoxia in DRG tissues were observed using hematoxylin-eosin staining and immunofluorescence. Western blotting and qPCR measured HIF-1α, GLUT1, PFKM, and PDK1 expression, while lactic acid and ATP levels in DRG tissue were also evaluated. Results: Gua Sha increased PWT and PWL, reduced serum and DRG inflammatory factors, improved back microcirculation, alleviated DRG hypoxia, and decreased HIF-1α and related signaling factors. It also lowered lactic acid and raised ATP levels. DMOG, a HIF-1α activator, reversed these effects. HIF-1α activation did not affect serum inflammatory factors but partially improved PWT. Inhibition of HIF-1α with YC-1 did not significantly differ from Gua Sha alone. Conclusion: HIF-1α-mediated metabolic reprogramming is a pathogenic mechanism in RIP. Gua Sha alleviates RIP by enhancing microcirculation, improving DRG hypoxia, inhibiting HIF-1α-mediated reprogramming, and reducing DRG sensitization and inflammation. This study provides insights into the mechanisms of Gua Sha's therapeutic effects in RIP.
Collapse
Affiliation(s)
- Haotian Ge
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shuxia Yan
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mingwan Yin
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yujie Gao
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiayi Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qin Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- TCM Nursing Intervention Laboratory of Chronic Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Guihua Xu
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Yang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
118
|
Lan Q, Ouyang A, Chen Y, Li Y, Zhong B, Deng S. Pain, lactate, and anesthetics: intertwined regulators of tumor metabolism and immunity. Front Oncol 2025; 15:1534300. [PMID: 40165895 PMCID: PMC11955471 DOI: 10.3389/fonc.2025.1534300] [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: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Patients with advanced cancer frequently endure severe pain, which substantially diminishes their quality of life and can adversely impact survival. Analgesia, a critical modality for alleviating such pain, is now under scrutiny for its potential role in cancer progression, a relationship whose underlying mechanisms remain obscure. Emerging evidence suggests that lactate, once considered a metabolic byproduct, actively participates in the malignant progression of cancer by modulating both metabolic and immunological pathways within the tumor microenvironment. Furthermore, lactate is implicated in the modulation of cancer-related pain, exerting effects through direct and indirect mechanisms. This review synthesizes current understanding of lactate's production, transport, and functional roles in tumor cells, encompassing the regulation of tumor metabolism, immunity, and progression. Additionally, we dissect the complex, bidirectional relationship between lactate and pain, and assess the impact of anesthetics on pain relief, lactate homeostasis, and tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Simin Deng
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
119
|
Arias CF, Acosta FJ, Bertocchini F, Fernández-Arias C. Redefining the role of hypoxia-inducible factors (HIFs) in oxygen homeostasis. Commun Biol 2025; 8:446. [PMID: 40089642 PMCID: PMC11910619 DOI: 10.1038/s42003-025-07896-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are key regulators of intracellular oxygen homeostasis. The marked increase in HIFs activity in hypoxia as compared to normoxia, together with their transcriptional control of primary metabolic pathways, motivated the widespread view of HIFs as responsible for the cell's metabolic adaptation to hypoxic stress. In this work, we suggest that this prevailing model of HIFs regulation is misleading. We propose an alternative model focused on understanding the dynamics of HIFs' activity within its physiological context. Our model suggests that HIFs would not respond to but rather prevent the onset of hypoxic stress by regulating the traffic of electrons between catabolic substrates and oxygen. The explanatory power of our approach is patent in its interpretation of the Warburg effect, the tendency of tumor cells to favor anaerobic metabolism over respiration, even in fully aerobic conditions. This puzzling behavior is currently considered as an anomalous metabolic deviation. Our model predicts the Warburg effect as the expected homeostatic response of tumor cells to the abnormal increase in metabolic demand that characterizes malignant phenotypes. This alternative perspective prompts a redefinition of HIFs' function and underscores the need to explicitly consider the cell's metabolic activity in understanding its responses to changes in oxygen availability.
Collapse
Affiliation(s)
- Clemente F Arias
- Grupo Interdisciplinar de Sistemas Complejos de Madrid (GISC), 28040, Madrid, Spain.
| | - Francisco J Acosta
- Departamento de Ecología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | - Cristina Fernández-Arias
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|
120
|
Cazeneuve C, Couret D, Lebeau G, Viranaicken W, Mathieu ME, Chouchou F. Protective Effect of Daily Physical Activity Against COVID-19 in a Young Adult Population on Reunion Island. Med Sci (Basel) 2025; 13:28. [PMID: 40137448 PMCID: PMC11944067 DOI: 10.3390/medsci13010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
The global fight against pandemics is a major public health issue. Epidemiological studies showed a reduced risk of the coronavirus disease 2019 (COVID-19) severity with the practice of regular physical activity (PA) in clinical populations. Here, we investigated the effect of PA against COVID-19 in a young general population. Methods: Two hundred ninety volunteers over 18 years old from Reunion Island responded to an online survey concerning sociodemographic, lifestyle and clinical information. Daily PA was studied using the International Physical Activity Questionnaire short version (IPAQ) and classified by overall score and intensities of PA. Results: Among 290 responders [179 women, median age = 27.5 years (interquartile range = 21.3 years)], 141 (48.6%) reported COVID-19 infection. Multivariate logistic analysis adjusted for age, sex, body mass index, chronic disease and alcohol consumption showed that the number of days per week of regular intense PA was independently associated with a low risk of COVID-19 infection [odds ratio (OR) 0.86; 95% confidence interval (CI) 0.24 to 0.99; p = 0.030], while regular moderate PA was not [OR 1.10; 95%CI 0.97 to 1.23; p = 0.137]. Conclusions: In a population of young adults, regular intense PA could offer a protective effect against COVID-19. Additional research is required to confirm this association in various viral infections and elucidate the fundamental mechanisms involved.
Collapse
Affiliation(s)
- Camille Cazeneuve
- Laboratoire d’IngéniéRIe de la Santé, du Sport et de l’Environnement (IRISSE, EA4075), UFR des Sciences de l’Homme et de l’Environnement, Université de La Réunion, 117 rue du General Ailleret, 97430 Le Tampon, La Réunion, France
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), Inserm UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, La Réunion, France
| | - David Couret
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), Inserm UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, La Réunion, France
| | - Gregorie Lebeau
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), Inserm UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, La Réunion, France
| | - Wildriss Viranaicken
- Diabète Athérothrombose Réunion Océan Indien (DéTROI), Inserm UMR 1188, Campus Santé de Terre Sainte, Université de La Réunion, 97410 Saint-Pierre, La Réunion, France
| | - Marie-Eve Mathieu
- School of Kinesiology and Physical Activity Sciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Centre de recherche Azrieli, CHU Saint-Justine, Montréal, QC H3T 1C5, Canada
| | - Florian Chouchou
- Laboratoire d’IngéniéRIe de la Santé, du Sport et de l’Environnement (IRISSE, EA4075), UFR des Sciences de l’Homme et de l’Environnement, Université de La Réunion, 117 rue du General Ailleret, 97430 Le Tampon, La Réunion, France
| |
Collapse
|
121
|
Llibre A, Kucuk S, Gope A, Certo M, Mauro C. Lactate: A key regulator of the immune response. Immunity 2025; 58:535-554. [PMID: 40073846 DOI: 10.1016/j.immuni.2025.02.008] [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: 11/22/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
Lactate, the end product of both anaerobic and aerobic glycolysis in proliferating and growing cells-with the latter process known as the Warburg effect-is historically considered a mere waste product of cell and tissue metabolism. However, research over the past ten years has unveiled multifaceted functions of lactate that critically shape and impact cellular biology. Beyond serving as a fuel source, lactate is now known to influence gene expression through histone modification and to function as a signaling molecule that impacts a wide range of cellular activities. These properties have been particularly studied in the context of both adaptive and innate immune responses. Here, we review the diverse roles of lactate in the regulation of the immune system during homeostasis and disease pathogenesis (including cancer, infection, cardiovascular diseases, and autoimmunity). Furthermore, we describe recently proposed therapeutic interventions for manipulating lactate metabolism in human diseases.
Collapse
Affiliation(s)
- Alba Llibre
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Salih Kucuk
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Atrayee Gope
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Michelangelo Certo
- College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
122
|
Chen S, Jiang Y, Zheng J, Li P, Liu M, Zhu Y, Zhu S, Chang S. Folate-targeted nanoparticles for glutamine metabolism inhibition enhance anti-tumor immunity and suppress tumor growth in ovarian cancer. J Control Release 2025; 379:89-104. [PMID: 39756690 DOI: 10.1016/j.jconrel.2024.12.073] [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: 08/07/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Ovarian cancer (OC) is a highly malignant gynecological tumor, and its effective treatment is frequently impeded by drug resistance and recurrent tumor growth. The reprogramming of glutamine metabolism in ovarian cancer is closely associated with tumor progression and the immunosuppressive tumor microenvironment. Recently, targeting metabolic reprogramming has emerged as a promising approach for cancer therapy. However, the application of such therapies is often constrained by their significant toxicity to normal tissues. In this study, we fabricated folate-targeted nanoparticles (FA-DCNPs) that co-encapsulate the glutamine metabolism inhibitor 6-diazo-5-oxo-L-norleucine (DON) and calcium carbonate (CaCO3). These nanoparticles alleviate damage to normal tissues by specifically targeting tumor cells via folate receptors (FOLR) mediation. Under acidic conditions, the FA-DCNPs release DON and Ca2+, generating a synergistic anti-tumor effect by impeding glutamine metabolism and inducing calcium overload. Additionally, FA-DCNPs target M2 phenotype tumor-associated macrophages (TAMs) via FOLR2, attenuating M2-TAMs activity. When partially phagocytosed by M0-TAMs, the nanoparticles restrict glutamate production, inhibiting polarization towards the M2 phenotype. This resulted in an increased proportion of M1-TAMs, thereby improving the tumor immune microenvironment. Our study explores a nanotherapeutic strategy that enhances the biosafety of anti-glutamine metabolism therapy through folate targeting, effectively suppresses tumor cell proliferation, and enhances the anti-tumor immune response.
Collapse
Affiliation(s)
- Shuning Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China; Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy, The Second Afliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China; State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yu Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Jiao Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging and Therapy, The Second Afliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Maoyu Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Shenyin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, People's Republic of China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China.
| |
Collapse
|
123
|
Soto CA, Lesch ML, Becker JL, Sharipol A, Khan A, Schafer XL, Becker MW, Munger JC, Frisch BJ. Elevated Lactate in the AML Bone Marrow Microenvironment Polarizes Leukemia-Associated Macrophages via GPR81 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.13.566874. [PMID: 39185193 PMCID: PMC11343108 DOI: 10.1101/2023.11.13.566874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Interactions between acute myeloid leukemia (AML) and the bone marrow microenvironment (BMME) are critical to leukemia progression and chemoresistance. In the solid tumor microenvironment, altered metabolite levels contribute to cancer progression. We performed a metabolomic analysis of AML patient bone marrow serum, revealing increased metabolites compared to age- and sex-matched controls. The most highly elevated metabolite in the AML BMME was lactate. Lactate signaling in solid tumors induces immunosuppressive tumor-associated macrophages and correlates with poor prognosis. This has not yet been studied in the leukemic BMME. Herein, we describe the role of lactate in the polarization of leukemia-associated macrophages (LAMs). Using a murine AML model of blast crisis chronic myelogenous leukemia (bcCML), we characterize the suppressive phenotype of LAMs by surface markers, transcriptomics, and cytokine profiling. Then, mice genetically lacking GPR81, the extracellular lactate receptor, were used to demonstrate GPR81 signaling as a mechanism of both the polarization of LAMs and the direct support of leukemia cells. Furthermore, elevated lactate diminished the function of hematopoietic progenitors and reduced stromal support for normal hematopoiesis. We report microenvironmental lactate as a mechanism of AML-induced immunosuppression and leukemic progression, thus identifying GPR81 signaling as an exciting and novel therapeutic target for treating this devastating disease.
Collapse
Affiliation(s)
- Celia A Soto
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Maggie L Lesch
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Jennifer L Becker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Azmeer Sharipol
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| | - Amal Khan
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Xenia L Schafer
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Joshua C Munger
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, NY, USA
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY, USA
| | - Benjamin J Frisch
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester School of Medicine, Rochester, NY, USA
| |
Collapse
|
124
|
Zhang F, Pan Z, Wu J, Huang Y. Relationship between MRI features and HIF-1α, GLUT1 and Ki-67 expression in pituitary adenoma with cystic degeneration. BMC Med Imaging 2025; 25:76. [PMID: 40050749 PMCID: PMC11887064 DOI: 10.1186/s12880-025-01574-8] [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: 08/01/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Pituitary adenomas (PAs) are prevalent tumors that often exhibit ischemia, hypoxia, and cystic transformations, impacting their prognosis. The relationship between cystic degeneration in PAs and the expressions of hypoxia-inducible factor-1α (HIF-1α), glucose transporter 1 (GLUT1), and Ki-67 remains unclear. This study aims to analyze the correlation between MRI characteristics of cystic PA and the expression of these proteins. METHODS This is a retrospective analysis. A total of 74 patients with cystic PA and 30 PA patients without cystic degeneration were enrolled. Their MRI signs were analyzed. According to the T2WI signs of PA, they were divided into the fluid level group (n = 26), non-fluid level group (n = 48), and non-cyst group (n = 30). Immunohistochemistry was performed to evaluate the expression levels of HIF-lα, GLUT1, and Ki-67 protein. Univariate and multinomial logistic regression analyses were used to evaluate the factors affecting MRI signs of PA. Spearman correlation was also performed. RESULTS There was no significant difference in gender, age, and HIF-1α protein expression among the three groups. Significant differences were found in invasiveness (P = 0.008), GLUT1 (P < 0.001), and Ki-67 protein expression (P = 0.009) among the three groups. Pairwise comparisons revealed statistically significant differences in invasiveness, GLUT1, and Ki-67 protein expressions between the non-fluid level group and the non-cyst group. Furthermore, GLUT1 protein expression was significantly different between the non-fluid level group and the fluid level group. Notably, GLUT1 was identified as an independent factor for the non-fluid level cystic characteristics of PA. Additionally, GLUT1 was positively correlated with invasiveness and Ki-67. CONCLUSION The non-fluid level cystic PA has higher invasiveness and higher proliferation than fluid level cystic PA and non-cyst PA, which may be related to high glucose metabolism as indicated by GLUT1 expression.
Collapse
Affiliation(s)
- Fangfang Zhang
- Department of Endocrinology, Fuzhou First General Hospital Affiliated with Fujian Medical University, Fuzhou, China
| | - Zhenhong Pan
- Fujian University of Traditional Chinese Medicine Fuzhou General Hospital, Fuzhou, China
| | - Jianwu Wu
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, (The 900TH Hospital), Xi'erhuanbei Road, Fuzhou, 350025, Fujian, China.
- Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, China.
| | - Yinxing Huang
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, (The 900TH Hospital), Xi'erhuanbei Road, Fuzhou, 350025, Fujian, China.
- Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, China.
| |
Collapse
|
125
|
Wang Y, Shi Y, Hu X, Wang C. Targeting glycolysis in esophageal squamous cell carcinoma: single-cell and multi-omics insights for risk stratification and personalized therapy. Front Pharmacol 2025; 16:1559546. [PMID: 40115255 PMCID: PMC11922847 DOI: 10.3389/fphar.2025.1559546] [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: 01/13/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is closely linked to aberrant glycolytic metabolism, a hallmark of cancer progression, immune evasion, and therapy resistance. This study employs single-cell transcriptomics and multi-omics approaches to unravel glycolysis-mediated mechanisms in ESCC, with a focus on risk stratification and therapeutic opportunities. Methods Data from TCGA and GEO databases were integrated with single-cell RNA sequencing, bulk RNA sequencing, as well as clinical datasets to investigate glycolysis-associated cell subtypes and their clinical implications in ESCC. Analytical approaches encompassed cell subtype annotation, cell-cell communication network analysis, and gene regulatory network modeling. A glycolysis-related risk score model was built via non-negative matrix factorization (NMF) and Cox regression, and then experimentally verified through Western blotting. Drug sensitivity analyses were carried out to explore potential therapeutic strategies. Results Single-cell analysis identified epithelial cells as the dominant glycolysis-active subtype, and tumor tissues showed significantly higher glycolytic activity than adjacent normal tissues. Among malignant epithelial subpopulations, IGFBP3+Epi (IGFBP3-expressing epithelial cells) and LHX9+Epi (LHX9-expressing epithelial cells) had elevated glycolysis levels, which correlated with poor prognosis, immune suppression, and changes in the tumor microenvironment. The seven-gene glycolysis-based risk score model divided patients into high- and low-risk groups, demonstrating strong prognostic performance. Drug sensitivity analysis showed high-risk patients were more responsive to Navitoclax as well as Rapamycin, but low-risk ones were more sensitive to Afatinib and Erlotinib, highlighting the model's usefulness in guiding personalized treatment. Conclusion This research emphasizes the crucial role of glycolysis in ESCC progression a well as immune modulation, offering a novel glycolysis-related risk score model with significant prognostic and therapeutic implications. These findings provide a basis for risk-based stratification and tailored therapeutic strategies, advancing precision medicine in ESCC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Anesthesia, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yunjie Shi
- Department of Anesthesia, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiao Hu
- Department of Anesthesia, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Chenfang Wang
- Department of Anesthesia, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
126
|
Di Patria L, Habel N, Olaso R, Fernandes R, Brenner C, Stefanovska B, Fromigue O. C-terminal binding protein-2 triggers CYR61-induced metastatic dissemination of osteosarcoma in a non-hypoxic microenvironment. J Exp Clin Cancer Res 2025; 44:83. [PMID: 40038783 PMCID: PMC11881356 DOI: 10.1186/s13046-025-03350-6] [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: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most prevalent cancer-related bone disease diagnosed in the pediatric age group. The rapid development of metastatic lesions and resistance to chemotherapy remain major mechanisms responsible for the failure of treatments and poor outcome. We established that the expression level of Cysteine-rich protein 61 (CYR61/CCN1) correlates to tumor neo-vascularization and dissemination in preclinical and clinical osteosarcoma samples. The aim of this study was to investigate the CYR61-related mechanisms leading to the acquisition of metastatic capacity by osteosarcoma cells. METHODS Transcriptomic data issued from RNA-seq were subjected to pathways and gene set enrichment analyses. Murine and human cell lines with overexpressed or downregulated C-terminal Binding protein 2 (CtBP2) were established by lentiviral transduction. Cell metabolic activity was assessed by Seahorse XF Analyzer; cell replication rate by BrdU incorporation assay; stemness by clonogenicity assay and RT-qPCR detection of markers; cell migration by wound healing assay and Boyden chambers system; cell invasion using Matrigel coated Boyden chambers or fluorescence microscopy of Matrigel embedded 3D spheroids. FFPE samples derived from syngeneic tumor cells grafts into BALB/c mice were analyzed by IHC. The protein interactome was predicted in silico using the STRING database. RESULTS GSEA revealed that CYR61 modulate the transcription process. The in vitro expression level of CtBP2 and Cyr61 correlated positively in a panel of osteosarcoma cell lines. In silico analysis of protein-protein interaction network revealed a link with stemness markers. Variations in CtBP2 expression levels influenced stemness markers expression levels, cell clonogenicity, cell migration, Matrix Metalloproteinase activity and cell invasion. Surprisingly, while induction of CtBP2 expression under CYR61 correlated with the metastatic dissemination process in vivo, it occurred only at the invasive front of tumors. Hypoxic conditions in central tumor region interfered with CtBP2 induction of expression. CONCLUSIONS Our findings identify for the first time that CtBP2 acts as a required critical inducing factor in the CYR61-related metastatic progression of osteosarcoma, by favoring cell migration and invasiveness. Moreover, we demonstrate that while CtBP2 is a downstream transcriptional target of CYR61 signaling cascade, it occurs only under non-hypoxic conditions. The present study suggests that CtBP2 may represent a potential pivotal target for therapeutic management of metastases spreading in osteosarcoma.
Collapse
Affiliation(s)
- Laura Di Patria
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Nadia Habel
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address : Centre de Traitement de L'Information Génétique (CTIG), INRAE, Jouy en Josas, France
| | - Robert Olaso
- Université Paris Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Romain Fernandes
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Catherine Brenner
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Bojana Stefanovska
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address: Department of Biochemistry and Structural Biology, Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Olivia Fromigue
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France.
| |
Collapse
|
127
|
Li Y, Xiao N, Wang Q, Liu B, Cui Y, Liu Y, Ji Y, Zheng M. Research on the mechanism of resistance exercise in promoting glucose metabolic shift to regulate muscle satellite cell proliferation in type 2 diabetic rats. Biochem Biophys Res Commun 2025; 751:151401. [PMID: 39923457 DOI: 10.1016/j.bbrc.2025.151401] [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: 12/16/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
Skeletal muscle atrophy is a common complication in patients with type 2 diabetes (T2D) and is associated with dysfunction of muscle satellite cells. The activation and proliferation of muscle satellite cells involve a switch in glucose metabolism, which is regulated by driving the acetylation of histones to control the expression of related genes. Studies have confirmed that resistance exercise can improve insulin resistance and activate muscle satellite cells, but the specific molecular mechanisms are not yet clear. This study aims to investigate whether resistance exercise can promote the proliferation of muscle satellite cells and improve muscle atrophy in type 2 diabetic rats by enhancing glucose metabolism in skeletal muscles. A T2D rat model was induced by combining a high-fat diet with streptozotocin injection. After 8 weeks of resistance exercise, the activity of key enzymes (Pyruvate Kinase, Phosphofructokinase, Pyruvate Dehydrogenase) in glucose metabolism in the skeletal muscles of T2D rats significantly increased, the expression of Sirtuin 1 (Sirt1) and Nicotin -amide Phosphoribosyltransferase (Nampt) in the skeletal muscles of the rats decreased, and the expression of acetylation of lysine 16 on histone H4 (H4K16ac) significantly increased, indicating an elevated level of the H4K16ac. The expression of paired box 7 (Pax7) and myogenic differentiation (MyoD) was significantly upregulated, indicating that exercise promoted the proliferation of muscle satellite cells. These results suggest that resistance exercise may promote glucose metabolism in skeletal muscles of T2D rats by regulating the activity of key enzymes in sugar metabolism, further regulating Sirt1-mediated histone H4K16ac, thereby promoting the proliferation of muscle satellite cells and improving muscle atrophy.
Collapse
MESH Headings
- Animals
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Satellite Cells, Skeletal Muscle/cytology
- Cell Proliferation
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/therapy
- Male
- Glucose/metabolism
- Physical Conditioning, Animal
- Rats
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/therapy
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Rats, Sprague-Dawley
- Sirtuin 1/metabolism
- Resistance Training
- Histones/metabolism
- Acetylation
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
Collapse
Affiliation(s)
- Ying Li
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Ningwen Xiao
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Qian Wang
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Bo Liu
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Ying Cui
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Yanyan Liu
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Ying Ji
- Harbin Sport University, Harbin, Heilongjiang, 150000, China
| | - Mi Zheng
- Harbin Sport University, Harbin, Heilongjiang, 150000, China.
| |
Collapse
|
128
|
Simoes RV, Henriques RN, Olesen JL, Cardoso BM, Fernandes FF, Monteiro MAV, Jespersen SN, Carvalho T, Shemesh N. Deuterium metabolic imaging phenotypes mouse glioblastoma heterogeneity through glucose turnover kinetics. eLife 2025; 13:RP100570. [PMID: 40035743 PMCID: PMC11879113 DOI: 10.7554/elife.100570] [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] [Indexed: 03/06/2025] Open
Abstract
Glioblastomas are aggressive brain tumors with dismal prognosis. One of the main bottlenecks for developing more effective therapies for glioblastoma stems from their histologic and molecular heterogeneity, leading to distinct tumor microenvironments and disease phenotypes. Effectively characterizing these features would improve the clinical management of glioblastoma. Glucose flux rates through glycolysis and mitochondrial oxidation have been recently shown to quantitatively depict glioblastoma proliferation in mouse models (GL261 and CT2A tumors) using dynamic glucose-enhanced (DGE) deuterium spectroscopy. However, the spatial features of tumor microenvironment phenotypes remain hitherto unresolved. Here, we develop a DGE Deuterium Metabolic Imaging (DMI) approach for profiling tumor microenvironments through glucose conversion kinetics. Using a multimodal combination of tumor mouse models, novel strategies for spectroscopic imaging and noise attenuation, and histopathological correlations, we show that tumor lactate turnover mirrors phenotype differences between GL261 and CT2A mouse glioblastoma, whereas recycling of the peritumoral glutamate-glutamine pool is a potential marker of invasion capacity in pooled cohorts, linked to secondary brain lesions. These findings were validated by histopathological characterization of each tumor, including cell density and proliferation, peritumoral invasion and distant migration, and immune cell infiltration. Our study bodes well for precision neuro-oncology, highlighting the importance of mapping glucose flux rates to better understand the metabolic heterogeneity of glioblastoma and its links to disease phenotypes.
Collapse
Affiliation(s)
- Rui Vasco Simoes
- Preclinical MRI, Champalimaud Research, Champalimaud FoundationLisbonPortugal
- Neuroengineering and Computational Neuroscience, Institute for Research and Innovation in Health (i3S)PortoPortugal
| | | | - Jonas L Olesen
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Physics and Astronomy, Aarhus UniversityAarhusDenmark
| | - Beatriz M Cardoso
- Preclinical MRI, Champalimaud Research, Champalimaud FoundationLisbonPortugal
| | | | - Mariana AV Monteiro
- Histopathology Platform, Champalimaud Research, Champalimaud FoundationLisbonPortugal
| | - Sune N Jespersen
- Center of Functionally Integrative Neuroscience (CFIN) and MINDLab, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Physics and Astronomy, Aarhus UniversityAarhusDenmark
| | - Tânia Carvalho
- Histopathology Platform, Champalimaud Research, Champalimaud FoundationLisbonPortugal
| | - Noam Shemesh
- Preclinical MRI, Champalimaud Research, Champalimaud FoundationLisbonPortugal
| |
Collapse
|
129
|
Gao Y, Sun M, Fu T, Wang Z, Jiang X, Yang L, Liang XG, Liu G, Tian Y, Yang F, Li J, Li Z, Li X, You Y, Ding C, Wang Y, Ma T, Zhang Z, Xu Z, Chen B, Yang Z. NOTCH, ERK, and SHH signaling respectively control the fate determination of cortical glia and olfactory bulb interneurons. Proc Natl Acad Sci U S A 2025; 122:e2416757122. [PMID: 39999176 PMCID: PMC11892625 DOI: 10.1073/pnas.2416757122] [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: 08/18/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
During cortical development, radial glial cells (neural stem cells) initially are neurogenic, generating intermediate progenitor cells that exclusively produce glutamatergic pyramidal neurons. Next, radial glial cells generate tripotential intermediate progenitor cells (Tri-IPCs) that give rise to cortical astrocytes and oligodendrocytes, and olfactory bulb interneurons. The molecular mechanisms underlying the transition from cortical neurogenesis to gliogenesis, and the subsequent fate determination of cortical astrocytes, oligodendrocytes, and olfactory bulb interneurons, remain unclear. Here, we report that extracellular signal-regulated kinase (ERK) signaling plays a fundamental role in promoting cortical gliogenesis and the generation of Tri-IPCs. Additionally, sonic hedgehog-smoothened-glioma-associated oncogene homolog (SHH-SMO-GLI) activator signaling has an auxiliary function to ERK during these processes. We further demonstrate that, from Tri-IPCs, NOTCH signaling is crucial for the fate determination of astrocytes, while ERK signaling plays a prominent role in oligodendrocyte fate specification, and SHH signaling is required for the fate determination of olfactory bulb interneurons. We provide evidence suggesting that this mechanism is conserved in both mice and humans. Finally, we propose a unifying principle of mammalian cortical gliogenesis.
Collapse
Affiliation(s)
- Yanjing Gao
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Mengge Sun
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Tongye Fu
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Ziwu Wang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Xin Jiang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Lin Yang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Xiaoyi G. Liang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA95064
| | - Guoping Liu
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Yu Tian
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Feihong Yang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Jialin Li
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Zhenmeiyu Li
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Xiaosu Li
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Yan You
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Chaoqiong Ding
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu610041, China
| | - Tong Ma
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Zhuangzhi Zhang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Zhejun Xu
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA95064
| | - Zhengang Yang
- Department of Neurology, Zhongshan Hospital, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| |
Collapse
|
130
|
Chen ZH, Cao SH, Ren ZY, Zhang T, Jiang HM, Hu ZK, Dong LH. Lactate Dehydrogenase A Crotonylation and Mono-Ubiquitination Maintains Vascular Smooth Muscle Cell Growth and Migration and Promotes Neointima Hyperplasia. J Am Heart Assoc 2025; 14:e036377. [PMID: 40028887 DOI: 10.1161/jaha.124.036377] [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: 05/01/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Phenotypic plasticity of vascular smooth muscle cells (VSMCs) is believed to be a key factor in neointima hyperplasia, which is the pathological basis of vascular remodeling diseases. LDHA (lactate dehydrogenase A) has been demonstrated to promote the proliferation and migration of VSMCs. However, the mechanism is still unclear. METHODS AND RESULTS LDHA ubiquitination and crotonylation in VSMCs were predicted by modified omics and proteomic analysis and were verified by immunoprecipitation. Lysine mutants of LDHA were conducted to determine the specific modified sites. Immunofluorescent staining, cell growth and migration assays, lactate production, immunobloting, adenovirus transduction, LDHA tetramerization, and mitochondrial extraction assays were performed to determine the molecular mechanism. LDHA expression, crotonylation, and ubiquitination in vivo were observed in the carotid arteries of ligation injury mice. We showed that the expression, crotonylation, and mono-ubiquitination of LDHA is upregulated in PDGF-BB (platelet-derived growth factor-BB)-induced proliferative VSMCs and ligation-induced neointima. LDHA is crotonylated at lysine 5 and is mono-ubiquitinated at K76. Crotonylation at lysine 5 activates LDHA through tetramer formation to enhance lactate production and VSMC growth. Mono-ubiquitination at K76 induces the translocation of LDHA into mitochondria, which promotes mitochondria fission and subsequent formation of lamellipodia and podosomes, thereby enhancing VSMC migration and growth. Furthermore, deletion of LDHA K5 crotonylation or K76 mono-ubiquitination decreases ligation-induced neointima formation. CONCLUSIONS Our study reveals a novel mechanism that combines VSMC metabolic reprogramming and vascular remodeling. Inhibition of LDHA K5 crotonylation or K76 mono-ubiquitination may be a promising approach for the therapy of vascular remodeling diseases.
Collapse
Affiliation(s)
- Zhi-Huan Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
- Hebei Special Education Collaborative Innovation Center School of Special Education, Handan University Handan China
| | - Shan-Hu Cao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Zhi-Yan Ren
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Ting Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Han-Mei Jiang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Zhao-Kun Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| | - Li-Hua Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Cardiovascular Medical Science Center, Key Laboratory of Vascular Biology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education Hebei Medical University Shijiazhuang People's Republic of China
| |
Collapse
|
131
|
Zhu K, Jin Y, Liu W, Wen C, Zheng X, Li Z, Chen Y, Niu Y, Pan W, Jiang Y, Jin Y. Clinical Investigations and Therapeutic Perspectives on Metabolic Syndrome following Kidney Transplantation. Kidney Blood Press Res 2025; 50:232-239. [PMID: 40037303 DOI: 10.1159/000545032] [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: 10/08/2024] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Kidney transplantation was an effective method for treating chronic kidney failure via transplanting a healthy kidney from a donor to a patient with the loss of kidney function. However, clinical studies revealed that the posttransplantation status of patients was associated with a substantial aggregation of risk factors contributing to metabolic syndrome. SUMMARY This article provided a comprehensive review of the current researches on metabolic syndrome after kidney transplantation, and the latest advances in the interaction between metabolism and immune cells were also covered. KEY MESSAGES Our aim was to identify and intervene high-risk recipients in time and thus improving the prognosis of recipients.
Collapse
Affiliation(s)
- Kejing Zhu
- Organ Transplantation Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yuji Jin
- School of Basic Medical Sciences, Jilin Medical University, Jilin, China
| | - Weijian Liu
- School of Basic Medical Sciences, Jilin Medical University, Jilin, China
| | - Cheng Wen
- School of Clinical Medicine, Jilin Medical University, Jilin, China
| | - Xinrui Zheng
- School of Clinical Medicine, Jilin Medical University, Jilin, China
| | - Zhixiong Li
- School of Clinical Medicine, Jilin Medical University, Jilin, China
| | - Yunjian Chen
- School of Clinical Medicine, Jilin Medical University, Jilin, China
| | - Yulin Niu
- Organ Transplantation Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Pan
- Guizhou Prenatal Diagnosis Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Jiang
- School of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Yingji Jin
- Department of Dermatology, Yanbian University Hospital, Yanji City, China
| |
Collapse
|
132
|
Verma N, Tiwari G, Khanna A, Mishra VK, Yadav Y, Malviya M, Sagar R. Molecular Design, Synthesis and Anti-cancer Activity of Novel Pyrazolo[3,4-b]pyridine-based Glycohybrid Molecules. Bioorg Chem 2025; 156:108161. [PMID: 39848166 DOI: 10.1016/j.bioorg.2025.108161] [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: 10/09/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
Molecular hybridization is an emerging strategy in medicinal chemistry for designing new bioactive molecules that link pharmacophores covalently and shows synergistic enhanced properties. Herein, we have developed pyrazolo[3,4-b]pyridine-based new glycohybrids considering the Warburg effect. A microwave-assisted, copper-catalyzed efficient synthesis of new triazole-linked glycohybrids based on pyrazolo[3,4-b]pyridines scaffold was achieved successfully in high yields with inherent stereochemical diversity from d-glucose, d-galactose, and d-mannose. The twenty-three distinct new glycohybrids, incorporating various electron-donating and electron-withdrawing groups with stereochemical diversities, were prepared using developed synthetic protocol. This efficient synthesis significantly reduced reaction time and furnished products with high isolated yields, showcasing its potential for glycohybrids synthesis. In-vitro study revealed that among the synthesized glycohybrids, compound 8e emerged as a potential compound against MDA-MB231 (SI > 31) and MCF-7 (SI > 434) with an IC50 value of 19.58 µM and 1.42 µM respectively. The molecular docking study predicts the binding interaction of the chemical probe with the target protein HCK. The enzyme inhibition assay revealed that compound 8e is having strong inhibitory potency against HCK enzyme. This article highlights the synthetic utility of this strategy and the potential applications of these newly designed and prepared glycohybrids.
Collapse
Affiliation(s)
- Neetu Verma
- Department of Chemistry, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Ghanshyam Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Vinay Kumar Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Yogesh Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Manisha Malviya
- Department of Chemistry, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India.
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India; Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
133
|
Yang L, Shi W, Li D, Shen Y, Li N, Meng Z. Study on the mechanism of 17-Hydroxy-jolkinolide B on anaplastic thyroid cancer cell. Am J Med Sci 2025; 369:405-412. [PMID: 39326738 DOI: 10.1016/j.amjms.2024.09.004] [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: 04/19/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) has a dismal prognosis, and the optimal treatment has not yet been confirmed. Euphorbia fischeriana Steud has been proven to exhibit pharmacological properties, including various antitumor effects, that can be used to treat numerous diseases and has been used to treat cancer. 17-Hydroxy-jolkinolide B (17-HJB) is one of the major diterpenoids produced from plants, but little research has investigated how it affects cancer. METHODS MTT assays, glucose and lactate concentration detection, Annexin V-FITC detection via cytometry, and Western blotting were performed to research the mechanism of 17-HJB. RESULTS Cell viability was inhibited in a concentration-dependent manner after 17-HJB treatment. 17-HJB inhibited glucose consumption and lactate production, and the expression of the glucose transporter GLUT1 and proteins associated with glycolysis, HK2, PFK1, and PKM2, was significantly downregulated. 17-HJB induced apoptosis, and the expression of signaling proteins related to apoptosis, such as Caspase-3 and cleaved Caspase-3, was upregulated. In vivo, 17-HJB effectively inhibited the growth of ATC tumors. The results of the expression of glycolysis-related enzyme proteins and apoptosis signaling proteins were consistent with those in vitro. CONCLUSIONS 17-HJB inhibited the growth of ATCs both in vivo and in vitro. The mechanism may be related to the effects on glucose metabolism and the inhibition of aerobic glycolysis. 17-HJB also induced ATC apoptosis.
Collapse
Affiliation(s)
- Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Wanying Shi
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard KarlsD University of Tuebingen, Tuebingen 72076, Germany
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Yiming Shen
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
134
|
Pan X, Cracan V. Translocation renal cell carcinoma says no to the Warburg effect. Nat Metab 2025; 7:438-440. [PMID: 39915637 PMCID: PMC11949694 DOI: 10.1038/s42255-025-01216-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
A new study reveals that in drastic contrast to other cancer types, translocation renal cell carcinoma (tRCC) is transcriptionally rewired towards an oxidative phosphorylation (OXPHOS) state, which renders tRCC vulnerable to interventions that promote NADH-reductive stress, highlighting how the maintenance of the optimal redox state in cancer can be therapeutically exploited.
Collapse
Affiliation(s)
- Xingxiu Pan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA
| | - Valentin Cracan
- Laboratory of Redox Biology and Metabolism, Scintillon Institute, San Diego, CA, USA.
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
135
|
Chopra A, Feldman M, Levy D. Orchestrating epigenetics: a comprehensive review of the methyltransferase SETD6. Exp Mol Med 2025; 57:533-544. [PMID: 40102573 PMCID: PMC11958702 DOI: 10.1038/s12276-025-01423-2] [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: 09/03/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 03/20/2025] Open
Abstract
Transcription is regulated by an intricate and extensive network of regulatory factors that impinge upon target genes. This process involves crosstalk between a plethora of factors that include chromatin structure, transcription factors and posttranslational modifications (PTMs). Among PTMs, lysine methylation has emerged as a key transcription regulatory PTM that occurs on histone and non-histone proteins, and several enzymatic regulators of lysine methylation are attractive targets for disease intervention. SET domain-containing protein 6 (SETD6) is a mono-methyltransferase that promotes the methylation of multiple transcription factors and other proteins involved in the regulation of gene expression programs. Many of these SETD6 substrates, such as the canonical SETD6 substrate RELA, are linked to cellular pathways that are highly relevant to human health and disease. Furthermore, SETD6 regulates numerous cancerous phenotypes and guards cancer cells from apoptosis. In the past 15 years, our knowledge of SETD6 substrate methylation and the biological roles of this enzyme has grown immensely. Here we provide a comprehensive overview of SETD6 that will enhance our understanding of this enzyme's role in chromatin and in selective transcriptional control, the contextual biological roles of this enzyme, and the molecular mechanisms and pathways in which SETD6 is involved, and we highlight the major trends in the SETD6 field.
Collapse
Affiliation(s)
- Anand Chopra
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Michal Feldman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
| | - Dan Levy
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
136
|
Bathe OF. Tumor metabolism as a factor affecting diversity in cancer cachexia. Am J Physiol Cell Physiol 2025; 328:C908-C920. [PMID: 39870605 DOI: 10.1152/ajpcell.00677.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025]
Abstract
Cancer cachexia is a multifaceted metabolic syndrome characterized by muscle wasting, fat redistribution, and metabolic dysregulation, commonly associated with advanced cancer but sometimes also evident in early-stage disease. More subtle body composition changes have also been reported in association with cancer, including sarcopenia, myosteatosis, and increased fat radiodensity. Emerging evidence reveals that body composition changes including sarcopenia, myosteatosis, and increased fat radiodensity, arise from distinct biological mechanisms and significantly impact survival outcomes. Importantly, these features often occur independently, with their combined presence exacerbating poor prognoses. Tumor plays a pivotal role in driving these host changes, either by acting as a metabolic parasite or by releasing mediators that disrupt normal tissue function. This review explores the diversity of tumor metabolism. It highlights the potential for tumor-specific metabolic phenotypes to influence systemic effects, including fat redistribution and sarcopenia. Addressing this tumor-host metabolic interplay requires personalized approaches that disrupt tumor metabolism while preserving host health. Promising strategies include targeted pharmacological interventions and anticachexia agents like growth differentiation factor 15 (GDF-15) inhibitors. Nutritional modifications such as ketogenic diets and omega-3 fatty acid supplementation also merit further investigation. In addition to preserving muscle, these therapies will need to be evaluated for their capability to improve survival and quality of life. This review underscores the need for further research into tumor-driven metabolic effects on the host and the development of integrative treatment strategies to address the interconnected challenges of cancer progression and cachexia.
Collapse
Affiliation(s)
- Oliver F Bathe
- Department of Surgery and Oncology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
137
|
Weng X, Gonzalez M, Angelia J, Piroozmand S, Jamehdor S, Behrooz AB, Latifi-Navid H, Ahmadi M, Pecic S. Lipidomics-driven drug discovery and delivery strategies in glioblastoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167637. [PMID: 39722408 DOI: 10.1016/j.bbadis.2024.167637] [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: 09/28/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
With few viable treatment options, glioblastoma (GBM) is still one of the most aggressive and deadly types of brain cancer. Recent developments in lipidomics have demonstrated the potential of lipid metabolism as a therapeutic target in GBM. The thorough examination of lipids in biological systems, or lipidomics, is essential to comprehending the changed lipid profiles found in GBM, which are linked to the tumor's ability to grow, survive, and resist treatment. The use of lipidomics in drug delivery and discovery is examined in this study, focusing on how it may be used to find new biomarkers, create multi-target directed ligands, and improve drug delivery systems. We also cover the use of FDA-approved medications, clinical trials that use lipid-targeted medicines, and the integration of lipidomics with other omics technologies. This study emphasizes lipidomics as a possible tool in developing more effective treatment methods for GBM by exploring various lipid-centric techniques.
Collapse
Affiliation(s)
- Xiaohui Weng
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Michael Gonzalez
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Jeannes Angelia
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States.
| |
Collapse
|
138
|
Rho H, Hay N. Protein lactylation in cancer: mechanisms and potential therapeutic implications. Exp Mol Med 2025; 57:545-553. [PMID: 40128358 PMCID: PMC11958728 DOI: 10.1038/s12276-025-01410-7] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 03/26/2025] Open
Abstract
Increased glycolysis, which leads to high lactate production, is a common feature of cancer cells. Recent evidence suggests that lactate plays a role in the post-translational modification of histone and nonhistone proteins via lactylation. In contrast to genetic mutations, lactylation in cancer cells is reversible. Thus, reversing lactylation can be exploited as a pharmacological intervention for various cancers. Here we discuss recent advances in histone and nonhistone lactylation in cancer, including L-, D- and S-lactylation, as well as alanyl-tRNA synthetase as a novel lactyltransferase. We also discuss potential approaches for targeting lactylation as a therapeutic opportunity in cancer treatment.
Collapse
Affiliation(s)
- Hyunsoo Rho
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea.
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
139
|
Lin DW, Carranza FG, Borrego S, Lauinger L, Dantas de Paula L, Pulipelli HR, Andronicos A, Hertel KJ, Kaiser P. Nutrient control of splice site selection contributes to methionine addiction of cancer. Mol Metab 2025; 93:102103. [PMID: 39862967 PMCID: PMC11834112 DOI: 10.1016/j.molmet.2025.102103] [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: 10/08/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Many cancer cells depend on exogenous methionine for proliferation, whereas non-tumorigenic cells can divide in media supplemented with the metabolic precursor homocysteine. This phenomenon is known as methionine dependence of cancer or methionine addiction. The underlying mechanisms driving this cancer-specific metabolic addiction are poorly understood. Here we find that methionine dependence is associated with severe dysregulation of pre-mRNA splicing. METHODS We used triple-negative breast cancer cells and their methionine-independent derivatives R8 to compare RNA expression profiles in methionine and homocysteine growth media. The data set was also analyzed for alternative splicing. RESULTS When tumorigenic cells were cultured in homocysteine medium, cancer cells failed to efficiently methylate the spliceosomal snRNP component SmD1, which resulted in reduced binding to the Survival-of-Motor-Neuron protein SMN leading to aberrant splicing. These effects were specific for cancer cells as neither Sm protein methylation nor splicing fidelity was affected when non-tumorigenic cells were cultured in homocysteine medium. Sm protein methylation is catalyzed by Protein Arginine Methyl Transferase 5 (Prmt5). Reducing methionine concentrations in the culture medium sensitized cancer cells to Prmt5 inhibition supporting a mechanistic link between methionine dependence of cancer and splicing. CONCLUSIONS Our results link nutritional demands to splicing changes and thereby provide a link between the cancer-specific metabolic phenomenon, described as methionine addiction over 40 years ago, with a defined cellular pathway that contributes to cancer cell proliferation.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Francisco G Carranza
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA
| | - Stacey Borrego
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Linda Lauinger
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Lucas Dantas de Paula
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Harika R Pulipelli
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA
| | - Anna Andronicos
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA
| | - Klemens J Hertel
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, USA.
| | - Peter Kaiser
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, USA.
| |
Collapse
|
140
|
Fiorentino F, Fabbrizi E, Mai A, Rotili D. Activation and inhibition of sirtuins: From bench to bedside. Med Res Rev 2025; 45:484-560. [PMID: 39215785 PMCID: PMC11796339 DOI: 10.1002/med.22076] [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: 05/25/2022] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
The sirtuin family comprises seven NAD+-dependent enzymes which catalyze protein lysine deacylation and mono ADP-ribosylation. Sirtuins act as central regulators of genomic stability and gene expression and control key processes, including energetic metabolism, cell cycle, differentiation, apoptosis, and aging. As a result, all sirtuins play critical roles in cellular homeostasis and organism wellness, and their dysregulation has been linked to metabolic, cardiovascular, and neurological diseases. Furthermore, sirtuins have shown dichotomous roles in cancer, acting as context-dependent tumor suppressors or promoters. Given their central role in different cellular processes, sirtuins have attracted increasing research interest aimed at developing both activators and inhibitors. Indeed, sirtuin modulation may have therapeutic effects in many age-related diseases, including diabetes, cardiovascular and neurodegenerative disorders, and cancer. Moreover, isoform selective modulators may increase our knowledge of sirtuin biology and aid to develop better therapies. Through this review, we provide critical insights into sirtuin pharmacology and illustrate their enzymatic activities and biological functions. Furthermore, we outline the most relevant sirtuin modulators in terms of their modes of action, structure-activity relationships, pharmacological effects, and clinical applications.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Emanuele Fabbrizi
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Antonello Mai
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
- Pasteur Institute, Cenci‐Bolognetti FoundationSapienza University of RomeRomeItaly
| | - Dante Rotili
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| |
Collapse
|
141
|
Yang Z, Su W, Zhang Q, Niu L, Feng B, Zhang Y, Huang F, He J, Zhou Q, Zhou X, Ma L, Zhou J, Wang Y, Xiong W, Xiang J, Hu Z, Zhan Q, Yao B. Lactylation of HDAC1 Confers Resistance to Ferroptosis in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408845. [PMID: 39888307 PMCID: PMC11947995 DOI: 10.1002/advs.202408845] [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: 07/30/2024] [Revised: 12/07/2024] [Indexed: 02/01/2025]
Abstract
Colorectal cancer (CRC) is highly resistant to ferroptosis, which hinders the application of anti-ferroptosis therapy. Through drug screening, it is found that histone deacetylase inhibitor (HDACi) significantly sensitized CRC to ferroptosis. The combination of HDACi and ferroptosis inducers synergically suppresses CRC growth both in vivo and in vitro. Mechanically, HDACi reduces ferroptosis suppressor protein (FSP1) by promoting its mRNA degradation. Specifically, it is confirmed that HDACi specifically targets HDAC1 and promotes the H3K27ac modification of fat mass- and obesity-associated gene (FTO) and AlkB Homolog 5, RNA Demethylase (ALKBH5), which results in significant activation of FTO and ALKBH5. The activation of FTO and ALKBH5 reduces N6-methyladenosine (m6A) modification on FSP1 mRNA, leading to its degradation. Crucially, lactylation of HDAC1K412 is essential for ferroptosis regulation. Both Vorinostat (SAHA) and Trichostatin A (TSA) notably diminish HDAC1K412 lactylation in comparison to other HDAC1 inhibitors, exhibiting a consistent trend of increasing susceptibility to ferroptosis. In conclusion, the research reveals that HDACi decreases HDAC1K412 lactylation to sensitize CRC to ferroptosis and that the combination of HDACi and ferroptosis inducers can be a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Wei Su
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Qinglin Zhang
- Departments of GastroenterologyWuxi People's Hospital Affiliated to Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsu214043China
| | - Lili Niu
- Department of Integrative MedicineShanghai Pulmonary HospitalTongji University Medical School Cancer InstituteTongji University School of MedicineShanghai200433China
| | - Baijie Feng
- Department of Medical OncologyShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Yu Zhang
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Feng Huang
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Jiaxin He
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Qinyao Zhou
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Xin Zhou
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Longjun Ma
- Department of EpidemiologySchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Jingwan Zhou
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Yuanrong Wang
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Wenjing Xiong
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Jun Xiang
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Zhilin Hu
- Department of ImmunologyKey Laboratory of Immune Microenvironment and DiseaseThe School of Basic Medicine; Department of laboratory medicine, the first affiliated hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing211166China
| | - Qiang Zhan
- Departments of GastroenterologyWuxi People's Hospital Affiliated to Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsu214043China
| | - Bing Yao
- Department of General Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine; National Experimental Teaching Center of Basic Medical Science, Department of Medical GeneticsSchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
- State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine; NHC Key Laboratory of Antibody Technique, Jiangsu Province Engineering Research Center of Antibody DrugNanjing Medical UniversityNanjing211166China
| |
Collapse
|
142
|
Lyu X, Yu Y, Jiang Y, Li Z, Qiao Q. The role of mitochondria transfer in cancer biological behavior, the immune system and therapeutic resistance. J Pharm Anal 2025; 15:101141. [PMID: 40115812 PMCID: PMC11925581 DOI: 10.1016/j.jpha.2024.101141] [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: 06/21/2024] [Revised: 10/14/2024] [Accepted: 11/03/2024] [Indexed: 03/23/2025] Open
Abstract
Mitochondria play a crucial role as organelles, managing several physiological processes such as redox balance, cell metabolism, and energy synthesis. Initially, the assumption was that mitochondria primarily resided in the host cells and could exclusively transmit from oocytes to offspring by a mechanism known as vertical inheritance of mitochondria. Recent scholarly works, however, suggest that certain cell types transmit their mitochondria to other developmental cell types via a mechanism referred to as intercellular or horizontal mitochondrial transfer. This review details the process of which mitochondria are transferred across cells and explains the impact of mitochondrial transfer between cells on the efficacy and functionality of cancer cells in various cancer forms. Specifically, we review the role of mitochondria transfer in regulating cellular metabolism restoration, excess reactive oxygen species (ROS) generation, proliferation, invasion, metastasis, mitophagy activation, mitochondrial DNA (mtDNA) inheritance, immune system modulation and therapeutic resistance in cancer. Additionally, we highlight the possibility of using intercellular mitochondria transfer as a therapeutic approach to treat cancer and enhance the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Xintong Lyu
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yangyang Yu
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yuanjun Jiang
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Zhiyuan Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110001, China
| | - Qiao Qiao
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
143
|
Maity A, Maidantchik VD, Weidenfeld K, Larisch S, Barkan D, Haick H. Chemical Tomography of Cancer Organoids and Cyto-Proteo-Genomic Development Stages Through Chemical Communication Signals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413017. [PMID: 39935131 PMCID: PMC11938034 DOI: 10.1002/adma.202413017] [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: 08/31/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Organoids mimic human organ function, offering insights into development and disease. However, non-destructive, real-time monitoring is lacking, as traditional methods are often costly, destructive, and low-throughput. In this article, a non-destructive chemical tomographic strategy is presented for decoding cyto-proteo-genomics of organoid using volatile signaling molecules, hereby, Volatile Organic Compounds (VOCs), to indicate metabolic activity and development of organoids. Combining a hierarchical design of graphene-based sensor arrays with AI-driven analysis, this method maps VOC spatiotemporal distribution and generate detailed digital profiles of organoid morphology and proteo-genomic features. Lens- and label-free, it avoids phototoxicity, distortion, and environmental disruption. Results from testing organoids with the reported chemical tomography approach demonstrate effective differentiation between cyto-proteo-genomic profiles of normal and diseased states, particularly during dynamic transitions such as epithelial-mesenchymal transition (EMT). Additionally, the reported approach identifies key VOC-related biochemical pathways, metabolic markers, and pathways associated with cancerous transformations such as aromatic acid degradation and lipid metabolism. This real-time, non-destructive approach captures subtle genetic and structural variations with high sensitivity and specificity, providing a robust platform for multi-omics integration and advancing cancer biomarker discovery.
Collapse
Affiliation(s)
- Arnab Maity
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
| | - Vivian Darsa Maidantchik
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
| | - Keren Weidenfeld
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Sarit Larisch
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Dalit Barkan
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
- Life Science Technology (LiST) GroupDanube Private UniversityFakultät Medizin/Zahnmedizin, Steiner Landstraße 124Krems‐Stein3500Austria
| |
Collapse
|
144
|
Li X, Zhu XH, Li Y, Wang T, Zhang G, Wiesner HM, Liang ZP, Chen W. Quantitative mapping of key glucose metabolic rates in the human brain using dynamic deuterium magnetic resonance spectroscopic imaging. PNAS NEXUS 2025; 4:pgaf072. [PMID: 40109558 PMCID: PMC11922071 DOI: 10.1093/pnasnexus/pgaf072] [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: 10/23/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
Deuterium (2H) magnetic resonance spectroscopic imaging (DMRSI) is a newly developed technology for assessing glucose metabolism by simultaneously measuring deuterium-labeled glucose and its downstream metabolites (1) and has a potential to provide a powerful neurometabolic imaging tool for quantitative studies of cerebral glucose metabolism involving multiple metabolic pathways in the human brain. In this work, we developed a dynamic DMRSI method that combines advanced radiofrequency coil and postprocessing techniques to substantially improve the imaging signal-to-noise ratio for detecting deuterated metabolites and enable robust dynamic DMRSI of the human brain at 7 T with very high resolution (HR; 0.7 cc nominal voxel and 2.5 min/image) and whole-brain coverage. Utilizing this capability, we were able to map and differentiate metabolite contents and dynamics throughout the human brain following oral administration of deuterated glucose. Furthermore, by introducing a sophisticated kinetic model, we demonstrated that three key cerebral metabolic rates of glucose consumption (CMRGlc), lactate production (CMRLac), and tricarboxylic acid (TCA) cycle (V TCA), as well as the maximum apparent rate of forward glucose transport (T max) can be simultaneously imaged in the human brain through a single dynamic DMRSI measurement. The results clearly show that the glucose transport, neurotransmitter turnover, CMRGlc, and V TCA are significantly higher in gray matter than in white matter in the human brain; and the mean metabolic rates and their ratios measured in this study are consistent with the values reported in the literature. The HR dynamic DMRSI methodology presented herein is of great significance and value for the quantitative assessment of human brain glucose metabolism, aerobic glycolysis, and metabolic reprogramming under physiopathological conditions.
Collapse
Affiliation(s)
- Xin Li
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Xiao-Hong Zhu
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Yudu Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tao Wang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Guangle Zhang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Hannes M Wiesner
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wei Chen
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
145
|
Burban A, Tessier C, Larroquette M, Guyon J, Lubiato C, Pinglaut M, Toujas M, Galvis J, Dartigues B, Georget E, Luchman HA, Weiss S, Cappellen D, Nicot N, Klink B, Nikolski M, Brisson L, Mathivet T, Bikfalvi A, Daubon T, Sharanek A. Exploiting metabolic vulnerability in glioblastoma using a brain-penetrant drug with a safe profile. EMBO Mol Med 2025; 17:469-503. [PMID: 39901019 PMCID: PMC11903783 DOI: 10.1038/s44321-025-00195-6] [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: 03/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
Glioblastoma is one of the most treatment-resistant and lethal cancers, with a subset of self-renewing brain tumour stem cells (BTSCs), driving therapy resistance and relapse. Here, we report that mubritinib effectively impairs BTSC stemness and growth. Mechanistically, bioenergetic assays and rescue experiments showed that mubritinib targets complex I of the electron transport chain, thereby impairing BTSC self-renewal and proliferation. Gene expression profiling and Western blot analysis revealed that mubritinib disrupts the AMPK/p27Kip1 pathway, leading to cell-cycle impairment. By employing in vivo pharmacokinetic assays, we established that mubritinib crosses the blood-brain barrier. Using preclinical patient-derived and syngeneic models, we demonstrated that mubritinib delays glioblastoma progression and extends animal survival. Moreover, combining mubritinib with radiotherapy or chemotherapy offers survival advantage to animals. Notably, we showed that mubritinib alleviates hypoxia, thereby enhancing ROS generation, DNA damage, and apoptosis in tumours when combined with radiotherapy. Encouragingly, toxicological and behavioural studies revealed that mubritinib is well tolerated and spares normal cells. Our findings underscore the promising therapeutic potential of mubritinib, warranting its further exploration in clinic for glioblastoma therapy.
Collapse
Affiliation(s)
- Audrey Burban
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Cloe Tessier
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | | | - Joris Guyon
- CHU of Bordeaux, Service de Pharmacologie Médicale, Bordeaux, France
- University of Bordeaux, INSERM, BPH, U1219, Bordeaux, France
| | - Cloe Lubiato
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Mathis Pinglaut
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Maxime Toujas
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Johanna Galvis
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
| | - Benjamin Dartigues
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Emmanuelle Georget
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - H Artee Luchman
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - David Cappellen
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
| | - Barbara Klink
- LuxGen Genome Center, Luxembourg Institute of Health, Laboratoire national de santé, Dudelange, Luxembourg
- National Center of Genetics (NCG), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg, 1526, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Macha Nikolski
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France
- Bordeaux Bioinformatic Center CBiB, University of Bordeaux, Bordeaux, France
| | - Lucie Brisson
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Thomas Mathivet
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France
| | - Andreas Bikfalvi
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR5095, Bordeaux, France.
| | - Ahmad Sharanek
- University of Bordeaux, INSERM, UMR1312, BRIC, BoRdeaux Institute of onCology, Bordeaux, France.
| |
Collapse
|
146
|
Schmitz I. A special RELationship between sugar and tumor-infiltrating regulatory T cells. Cell Mol Immunol 2025; 22:330-332. [PMID: 39681605 PMCID: PMC11868515 DOI: 10.1038/s41423-024-01248-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Ingo Schmitz
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
147
|
Mohd Sahardi NFN, Priya M, Makpol S, Shafiee MN. Clinical translation of metabolomics markers in endometrial carcinoma. J Obstet Gynaecol Res 2025; 51:e16246. [PMID: 40015330 DOI: 10.1111/jog.16246] [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: 09/10/2024] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE This comprehensive review highlights the current research on metabolomics and the metabolic pathways involved in endometrial cancer (EC), offering potential non-invasive biomarkers for EC. METHODS The data was extracted from published manuscripts between 2015 and 2024 using the reputed search engine "Pubmed." All gathered data were organized into a single table, facilitating a comparison with earlier findings. RESULTS The results of this study revealed most metabolites identified in previous metabolomic research on EC are associated with lipid, glucose, and amino acid metabolism. CONCLUSION Therefore, understanding these metabolic pathway alterations in EC is crucial for improving diagnosis, prognosis, and treatments by specially targeting these metabolic pathways.
Collapse
Affiliation(s)
| | - Manishaa Priya
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohamad Nasir Shafiee
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
148
|
Luo Y, Zhang N, Ye J, Wang Z, Zhou X, Liu J, Cai J, Li C, Chen L. Unveiling lactylation modification: A new hope for cancer treatment. Biomed Pharmacother 2025; 184:117934. [PMID: 39986235 DOI: 10.1016/j.biopha.2025.117934] [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: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025] Open
Abstract
This review article delves into the multifaceted role of lactylation modification in antitumor therapy, revealing the complex interplay between lactylation modification and the tumor microenvironment (TME), metabolic reprogramming, gene expression, and immunotherapy. As an emerging epigenetic modification, lactylation has a significant impact on the metabolic pathways of tumor cells, immune evasion, gene expression regulation, and sensitivity to chemotherapy drugs. Studies have shown that lactylation modification significantly alters the development and therapeutic response of tumors by affecting metabolites in the TME, immune cell functions, and signaling pathways. In the field of immunotherapy, the regulatory role of lactylation modification provides a new perspective and potential targets for tumor treatment, including modulating the sensitivity of tumors to immunotherapy by affecting the expression of immune checkpoint molecules and the infiltration of immune cells. Moreover, research progress on lactylation modification in various types of tumors indicates that it may serve as a biomarker to predict patients' responses to chemotherapy and immunotherapy. Overall, research on lactylation modification provides a theoretical foundation for the development of new tumor treatment strategies and holds promise for improving patient prognosis and quality of life. Future research will further explore the application potential of lactylation modification in tumor therapy and how to improve treatment efficacy by targeting lactylation modification.
Collapse
Affiliation(s)
- Yuxiang Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Ning Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Chen Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi 330006, China; Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Jiangxi 330006, China; Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi 330006, China.
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Precision Oncology Medicine Center,The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China.
| |
Collapse
|
149
|
Guo Z, Xiao Y, Wu W, Zhe M, Yu P, Shakya S, Li Z, Xing F. Metal-organic framework-based smart stimuli-responsive drug delivery systems for cancer therapy: advances, challenges, and future perspectives. J Nanobiotechnology 2025; 23:157. [PMID: 40022098 PMCID: PMC11871784 DOI: 10.1186/s12951-025-03252-x] [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: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Cancer treatment is currently one of the most critical healthcare issues globally. A well-designed drug delivery system can precisely target tumor tissues, improve efficacy, and reduce damage to normal tissues. Stimuli-responsive drug delivery systems (SRDDSs) have shown promising application prospects. Intelligent nano drug delivery systems responsive to endogenous stimuli such as weak acidity, complex redox characteristics, hypoxia, active energy metabolism, as well as exogenous stimuli like high temperature, light, pressure, and magnetic fields are increasingly being applied in chemotherapy, radiotherapy, photothermal therapy, photodynamic therapy, and various other anticancer approaches. Metal-organic frameworks (MOFs) have become promising candidate materials for constructing SRDDSs due to their large surface area, tunable porosity and structure, ease of synthesis and modification, and good biocompatibility. This paper reviews the application of MOF-based SRDDSs in various modes of cancer therapy. It summarizes the key aspects, including the classification, synthesis, modifications, drug loading modes, stimuli-responsive mechanisms, and their roles in different cancer treatment modalities. Furthermore, we address the current challenges and summarize the potential applications of artificial intelligence in MOF synthesis. Finally, we propose strategies to enhance the efficacy and safety of MOF-based SRDDSs, ultimately aiming at facilitating their clinical translation.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiyun Yu
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
150
|
Quan C, Jiang X. The molecular mechanism underlying the human glucose facilitators inhibition. VITAMINS AND HORMONES 2025; 128:49-92. [PMID: 40097253 DOI: 10.1016/bs.vh.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose is the primary energy substrate and an essential precursor for cellular metabolism. Maintaining glucose homeostasis necessitates the presence of glucose transporters, as the hydrophilic nature of glucose prevents its passage across the cell membrane. The GLUT family is a crucial group of glucose transporters that facilitate glucose diffusion along the transmembrane glucose concentration gradient. Dysfunction in GLUTs is associated with diseases, such as GLUT1 deficiency syndrome, Fanconi-Bickel syndrome, and type 2 diabetes. Furthermore, elevated expression of GLUTs fuels aerobic glycolysis, known as the Warburg effect, in various types of cancers, making GLUT isoforms possible targets for antineoplastic therapies. To date, 30 GLUT and homolog structures have been released on the Protein Data Bank (PDB), showcasing multiple conformational and ligand-binding states. These structures elucidate the molecular mechanisms underlying substrate recognition, the alternating access cycle, and transport inhibition. Here, we summarize the current knowledge of human GLUTs and their role in cancer, highlighting recent advances in the structural characterization of GLUTs. We also compare the inhibition mechanisms of exofacial and endofacial GLUT inhibitors, providing insights into the design and optimization of GLUT inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Cantao Quan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China
| | - Xin Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China.
| |
Collapse
|