1
|
Duan Y, Liu Z, Wang Q, Zhang J, Liu J, Zhang Z, Li C. Targeting MYC: Multidimensional regulation and therapeutic strategies in oncology. Genes Dis 2025; 12:101435. [PMID: 40290126 PMCID: PMC12022651 DOI: 10.1016/j.gendis.2024.101435] [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: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 04/30/2025] Open
Abstract
MYC is dysregulated in approximately 70% of human cancers, strongly suggesting its essential function in cancer. MYC regulates many biological processes, such as cell cycle, metabolism, cellular senescence, apoptosis, angiogenesis, and immune escape. MYC plays a central role in carcinogenesis and is a key regulator of tumor development and drug resistance. Therefore, MYC is one of the most alluring therapeutic targets for developing cancer drugs. Although the search for direct inhibitors of MYC is challenging, MYC cannot simply be assumed to be undruggable. Targeting the MYC-MAX complex has been an effective method for directly targeting MYC. Alternatively, indirect targeting of MYC represents a more pragmatic therapeutic approach, mainly including inhibition of the transcriptional or translational processes of MYC, destabilization of the MYC protein, and blocking genes that are synthetically lethal with MYC overexpression. In this review, we delineate the multifaceted roles of MYC in cancer progression, highlighting a spectrum of therapeutic strategies and inhibitors for cancer therapy that target MYC, either directly or indirectly.
Collapse
Affiliation(s)
- Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiaxin Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziyi Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
2
|
Wang C, Liang W, Zhong J, Liu J, Zhou C, Ma C, Liao Y, Gao Y, Zhao J, He Y. m6A-mediated regulation of CPSF6 by METTL3 promotes oxaliplatin resistance in colorectal cancer through enhanced glycolysis. Cell Signal 2025; 130:111676. [PMID: 40010558 DOI: 10.1016/j.cellsig.2025.111676] [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: 10/02/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Oxaliplatin resistance poses a significant challenge in colorectal cancer (CRC) treatment. Recent studies have implicated CPSF6 in cancer progression and drug resistance, although its role in chemotherapy resistance and regulatory mechanisms is unclear. This study investigates CPSF6's involvement in oxaliplatin resistance in CRC and its regulation via m6A methylation by METTL3. We assessed CPSF6 expression in oxaliplatin-resistant (OxR) CRC cell lines (HT29-OxR and HCT116-OxR) compared to sensitive counterparts using qRT-PCR and Western blotting. CPSF6 was significantly upregulated in OxR cells, and its knockdown reduced cell viability, colony formation, and glycolytic activity while increasing apoptosis. m6A modification of CPSF6 mRNA was elevated in OxR cells, correlating with increased METTL3 expression. METTL3 knockdown decreased CPSF6 levels and m6A enrichment, enhancing mRNA degradation, while its overexpression stabilized CPSF6 mRNA, promoting resistance. Xenograft experiments showed that CPSF6 knockdown suppressed tumor growth and glycolysis. Thus, CPSF6 is identified as a mediator of oxaliplatin resistance in CRC, regulated by the METTL3/m6A axis, suggesting potential therapeutic targets to overcome resistance.
Collapse
Affiliation(s)
- Chengxing Wang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jietao Zhong
- Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China; Department of Gastroenterology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jiachen Liu
- Department of Nuclear Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong 510000, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Changyi Ma
- Department of Radiology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuehua Liao
- Department of Pathology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jinglin Zhao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| |
Collapse
|
3
|
Zeng Y, Tao Y, Du G, Huang T, Chen S, Fan L, Zhang N. Advances in the mechanisms of HIF-1α-enhanced tumor glycolysis and its relation to dedifferentiation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00024-0. [PMID: 40373959 DOI: 10.1016/j.pbiomolbio.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Metabolic reprogramming, a hallmark of malignancy, enables tumor cells to adapt to the harsh and dynamic tumor microenvironment (TME) by altering metabolic pathways. Hypoxia, prevalent in solid tumors, activates hypoxia inducible factor 1α (HIF-1α). HIF-1α drives metabolic reprogramming, enhancing glycolysis primarily through the Warburg effect to reduce oxygen dependence and facilitate tumor cell growth/proliferation. The above process is associated with accelerated tumor cell dedifferentiation and enhanced stemness, generating cancer stem cells (CSCs) which possesses the potential for self-renewal and differentiation that can differentiate into a wide range of subtypes of tumor cells and fuel tumor heterogeneity, metastasis, and recurrence, complicating therapy. This review examines the HIF-1α-glycolysis-dedifferentiation crosstalk mechanisms, expecting that indirect inhibition of HIF-1α by targeting metabolic enzymes, metabolites, or their signaling pathways will offer an effective therapeutic strategy to improve the cancer treatment outcomes.
Collapse
Affiliation(s)
- Yu Zeng
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Yonggang Tao
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Guotu Du
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Tianyu Huang
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Shicheng Chen
- Zunyi China, The Second Affiliated Hospital of Zunyi Medical University(2)
| | - Longmei Fan
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1)
| | - Neng Zhang
- Zunyi China, The Affiliated Hospital of Zunyi Medical University(1).
| |
Collapse
|
4
|
Berends AMA, Wardenaar R, van den Bos H, Tijhuis AE, Links TP, Feelders RA, Hofland LJ, Kruijff S, Pacak K, Spierings DCJ, Kerstens MN, Foijer F. Single-cell chromosome and bulk transcriptome analysis as a diagnostic tool to differentiate between localized and metastatic pheochromocytoma and sympathetic paraganglioma. Oncogene 2025:10.1038/s41388-025-03391-3. [PMID: 40319142 DOI: 10.1038/s41388-025-03391-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/16/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
Approximately 10-20% of patients with pheochromocytoma or sympathetic paraganglioma (PPGL) develop metastatic disease, most often as metachronous lesions. Unfortunately, there is a lack of accurate biomarkers that can predict the biologic behavior of a PPGL at the initial diagnosis. We investigated tumor samples from patients with PPGL and a diagnosis of either localized or metastatic disease with synchronous or metachronous metastases and performed a comprehensive molecular analysis through application of single-cell whole-genome sequencing and bulk transcriptome analysis, including variant detection analysis of RNA sequences. We found that PPGL displayed complex karyotypes with recurrent aneuploidies and substantial cell-to-cell karyotype variability, indicating ongoing chromosomal instability (CIN) in both localized and metastatic tumors. Transcriptome analysis on the other hand revealed several differences between localized and metastatic PPGL including TNFα and TGFβ signaling in metastatic PPGL that were already detectable in primary tumor samples of initially non-metastatic-appearing PPGLs that developed metachronous metastases. Altogether our findings indicate that while localized and metastatic PPGL in general have comparable genomic landscapes, they do show transcriptional differences that are already detectable in primary tumor PPGL before development of metastases. This finding could provide an important tool for improvement of patient stratification at initial diagnosis.
Collapse
Affiliation(s)
- Annika M A Berends
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hilda van den Bos
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andréa E Tijhuis
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Thera P Links
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Richard A Feelders
- Department of Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Schelto Kruijff
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD, USA
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel N Kerstens
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Zi R, Zhao X, Liu L, Wang Y, Zhang R, Bian Z, Jiang H, Liu T, Sun Y, Peng H, Wang X, Lu F, Zhang C, Zhang F, Qin Q, Liang H, Li J, Wei Z, Dong Y. Metabolic-Immune Suppression Mediated by the SIRT1-CX3CL1 Axis Induces Functional Enhancement of Regulatory T Cells in Colorectal Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404734. [PMID: 39783838 PMCID: PMC12061293 DOI: 10.1002/advs.202404734] [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] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/01/2024] [Indexed: 01/12/2025]
Abstract
Metabolic reprogramming of tumor cells dynamically reshapes the distribution of nutrients and signals in the tumor microenvironment (TME), affecting intercellular interactions and resulting in metabolic immune suppression. Increased glucose uptake and metabolism are characteristic of many tumors. Meanwhile, the progression of colorectal carcinoma (CRC) relies on lipid metabolism. Therefore, investigating the role of glucolipid metabolic reprogramming on tumor immunity contributes to identifying new targets for immune suppression intervention in CRC. Our previous work demonstrated that SIRT1 is the hub gene involved in glucolipid metabolic conversion in CRC. Here, it is found that upregulated SIRT1 in CRC cells increases Treg functionality by promoting the secretion of CX3CL1. The CX3CL1-CX3CR1 signaling activated transcription factors SATB1 and BTG2, promoting the differentiation of TCF7+ Treg cells into functionally enhanced TNFRSF9+ Treg cells. Multiplex immunofluorescence (mIHC) analysis of a CRC tissue microarray confirmed the promoting effect of CX3CL1 on Treg infiltration. Additionally, the therapeutic efficacy of CX3CR1 inhibitor monotherapy and combination therapy is validated with the PD-1 antibody in the humanized subcutaneous CRC mouse model. This study elucidates a potential mechanism that metabolic reprogramming of cancer cells collaborates with subsequent immunosuppression to promote CRC progression.
Collapse
Affiliation(s)
- Ruiyang Zi
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Xiang Zhao
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Limei Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized TreatmentCancer HospitalChongqing UniversityChongqing400038China
| | - Yijie Wang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Rui Zhang
- Department of Stem Cell and Regenerative MedicineSouthwest HospitalThird Military Medical University (Army Medical University)ChongQing400038China
| | - Zhiheng Bian
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Haoran Jiang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Taorui Liu
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Yixin Sun
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Han Peng
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Xuesong Wang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Fanghao Lu
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Chao Zhang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Fan Zhang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Qing Qin
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Houjie Liang
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Zhihao Wei
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
- Brain Research Center and State Key Laboratory of TraumaBurns, and Combined InjuryThird Military Medical UniversityChongqing400038China
| | - Yan Dong
- Department of Oncology and Southwest Cancer CenterSouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| |
Collapse
|
6
|
Filippi L, Innocenti F, Pascarella F, Scaramuzzo RT, Morganti R, Bagnoli P, Cammalleri M, Dal Monte M, Calvani M, Pini A. β 3-Adrenoceptor Agonism to Mimic the Biological Effects of Intrauterine Hypoxia: Taking Great Strides Toward a Pharmacological Artificial Placenta. Med Res Rev 2025; 45:842-866. [PMID: 39604126 PMCID: PMC11976384 DOI: 10.1002/med.22092] [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: 07/31/2024] [Revised: 10/24/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
At different stages of life, from embryonic to postnatal, varying oxygen concentrations modulate cellular gene expression by enhancing or repressing hypoxia-inducible transcription factors. During embryonic/fetal life, these genes encode proteins involved in adapting to a low-oxygen environment, including the induction of specific enzymes related to glycolytic metabolism, erythropoiesis, angiogenesis, and vasculogenesis. However, oxygen concentrations fluctuate during intrauterine life, enabling the induction of tissue-specific differentiation processes. Fetal well-being is thus closely linked to the physiological benefits of a dynamically hypoxic environment. Premature birth entails the precocious exposure of the immature fetus to a more oxygen-rich environment compared to the womb. As a result, preterm newborns face a condition of relative hyperoxia, which alters the postnatal development of organs and contributes to prematurity-related diseases. However, until recently, the molecular mechanism by which high oxygen tension alters normal fetal differentiation remained unclear. In this review, we discuss the research trajectory followed by our research group, which suggests that early exposure to a relatively hyperoxic environment may impair preterm neonates due to reduced expression of the β3-adrenoceptor. Additionally, we explore how these impairments could be prevented through the pharmacological stimulation of the remaining β3-adrenoceptors. Recent preclinical studies demonstrate that pharmacological stimulation of the β3-adrenoceptor can decouple exposure to hyperoxia from its harmful effects, offering a glimpse of the possibility to recreating the conditions typical of intrauterine life, even after premature birth.
Collapse
Affiliation(s)
- Luca Filippi
- Neonatology UnitAzienda Ospedaliero‐Universitaria PisanaPisaItaly
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | | | | | | | - Riccardo Morganti
- Section of StatisticsAzienda Ospedaliero‐Universitaria PisanaPisaItaly
| | - Paola Bagnoli
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Maurizio Cammalleri
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Massimo Dal Monte
- Department of Biology, Unit of General PhysiologyUniversity of PisaPisaItaly
| | - Maura Calvani
- Department of Pediatric Hematology‐OncologyMeyer Children's Hospital IRCCSFlorenceItaly
| | - Alessandro Pini
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| |
Collapse
|
7
|
Fan C, Fan J, Chen H, Lin S, Zhang D, Song J, Wang J, Wang Y, Han X, Yuan J. Switching the yeast metabolism via manipulation of sugar phosphorylation. Metab Eng 2025; 89:76-85. [PMID: 39988025 DOI: 10.1016/j.ymben.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: 09/02/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Saccharomyces cerevisiae predominantly ferments sugar to ethanol, irrespective of the presence of oxygen, which is known as the Crabtree-effect. Traditional methods rely on static controls of glycolytic flux to make S. cerevisiae Crabtree-negative, which are not favorable for future biomanufacturing applications. Considering native metabolic pathways typically harness dynamic regulatory networks, we therefore aim to develop an alternative strategy using dynamic regulation of the yeast central metabolism to generate Crabtree-negative S. cerevisiae. We report that manipulating a single step at sugar phosphorylation can alter the mode of yeast metabolism with an attenuated Crabtree-effect. By implementing catabolite-regulated sugar phosphorylation, the diauxic shift in budding yeast was effectively reduced. The Crabtree-attenuated metabolism in the engineered yeast was confirmed by multidimensional characterizations such as cell morphology, the measurements of sugar utilization rate and ethanol production, and transcriptomics. In addition, we demonstrated that the Crabtree-attenuated metabolism could substantially improve the mitochondrial synthesis of short branched-chain fatty acids from amino acid catabolism, and allow the synthesis and accumulation of retinaldehyde. Taken together, we present for the first time that manipulation of sugar phosphorylation can alter the mode of yeast metabolism, and the synthetic Crabtree-attenuated yeast factory established here might serve as a non-fermentative biomanufacturing chassis.
Collapse
Affiliation(s)
- Cong Fan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Jian Fan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Haofeng Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Shujin Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Danli Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Jingya Song
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Junyi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Yan Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Xiao Han
- College of Biological Science and Engineering, Fuzhou University, Fujian, 350108, China
| | - Jifeng Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China; Key Laboratory for Synthetic Biotechnology of Xiamen City, Xiamen University, Fujian, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
8
|
Tran RL, Li T, de la Cerda J, Schuler FW, Khaled AS, Pudakalakatti S, Bhattacharya PK, Sinharay S, Pagel MD. Potentiation of immune checkpoint blockade with a pH-sensitizer as monitored in two pre-clinical tumor models with acidoCEST MRI. Br J Cancer 2025; 132:744-753. [PMID: 39994445 PMCID: PMC11997056 DOI: 10.1038/s41416-025-02962-1] [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: 08/26/2023] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Tumor acidosis causes resistance to immune checkpoint blockade (ICB). We hypothesized that a "pH-sensitizer" can increase tumor extracellular pH (pHe) and improve tumor control following ICB. We also hypothesized that pHe measured with acidoCEST MRI can predict improved tumor control with ICB. METHODS We tested the effects of pH-sensitizers on proton efflux rate (PER), cytotoxicity, T cell activation, tumor immunogenicity, tumor growth and survival using 4T1 and B16-F10 tumor cells. We measured in vivo tumor pHe of 4T1 and B16-F10 models with acidoCEST MRI. RESULTS Among the pH-sensitizers tested, someprazole caused the greatest reduction in PER without exhibiting cytotoxicity or reducing T cell activation. Esomeprazole improved 4T1 tumor control with ICB administered one day after the pH-sensitizer. Tumor pHe positively correlated with TCF-1 + CD4 effector and CD8 T cell intratumoral frequencies and predicted improved 4T1 tumor control with ICB. For comparison, esomeprazole had a mild effect on B16-F10 tumor pHe, and worsened tumor control with ICB and increased intratumoral myeloid and dendritic cell (DC) frequencies. CONCLUSIONS A pH-sensitizer can improve tumor control with ICB, and acidoCEST MRI can be used to measure pHe and predict tumor control, but only in the 4T1 model and not the B16-F10 model.
Collapse
Affiliation(s)
- Renee L Tran
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Tianzhe Li
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jorge de la Cerda
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - F William Schuler
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Alia S Khaled
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sanhita Sinharay
- Centre for Biosystems Science & Engineering, Indian Institute of Science, Bangalore, India
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Zuo Y, Ren D, He H, Huang C, Zhu X. CircST6GALNAC6 Inhibits Glycolysis of Bladder Cancer by Regulating PRKN/HK1 Signaling Pathway. Mol Carcinog 2025; 64:870-882. [PMID: 39960214 DOI: 10.1002/mc.23894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 04/12/2025]
Abstract
Bladder cancer (BCa) is an aggressive malignancy of urinary system. Aerobic glycolysis refers to the phenomenon wherein cancer cells increase glucose consumption and produce lactic acid. Our study focused on the role and mechanism of circST6GALNAC6 in BCa glycolysis. The 24 h glucose intake was detected using flow cytometry. Lactic acid and ATP were detected in BCa cells utilizing commercially provided kits. Extracellular acidification rate was measured using Seahorse XF-96p Extracellular Flux Analyzer. Cell proliferation was determined using colony formation assay. RNA immunoprecipitation and co-immunoprecipitation experiments were adopted to validate molecular interactions. BALB/C nude mice were utilized to establish xenograft tumor model. CircST6GALNAC6 was decreased in BCa cells, and overexpression of circST6GALNAC6 inhibited glycolysis and proliferation of BCa cells. Additionally, overexpression of circST6GALNAC6 promoted the degradation of glycolytic regulatory protein HK1 and decreased its expression, and PRKN facilitated ubiquitination-related degradation of HK1. CircST6GALNAC6 enhanced the mRNA stability and expression of PRKN by recruiting FUS. Furthermore, the inhibitory impact of circST6GALNAC6 overexpression on glycolysis in BCa cells was reversed by PRKN knockdown. Finally, overexpression of circST6GALNAC6 suppressed tumor growth through increasing PRKN in nude mice. CircST6GALNAC6 suppressed glycolysis in BCa through FUS/PRKN/HK1 axis. Targeting circST6GALNAC6 holds promise as a novel approach for treating BCa.
Collapse
Affiliation(s)
- Yali Zuo
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Da Ren
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haiqing He
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Changkun Huang
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xuan Zhu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
10
|
Chen H, Ding H, Huang D, Wu S. GBE1 alleviates MPTP-induced PD symptoms in mice by enhancing glycolysis and oxidative phosphorylation. Brain Res 2025; 1859:149663. [PMID: 40316161 DOI: 10.1016/j.brainres.2025.149663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/20/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
In Parkinson's disease (PD), the disturbance of energy metabolism due to glucose metabolic reprogramming may be a critical factor contributing to neuronal degeneration and death. Glycolysis, as the core process of glucose metabolism, not only serves as a fundamental source of energy but also integrates various metabolic pathways. However, the precise role of alterations in glycolysis-related pathways in the progression of PD remains elusive. We compared and analysed datasets from human databases of patients with PD and healthy controls to identify differentially expressed genes associated with glycolysis. Using the least absolute shrinkage and selection operator regression method and multivariate logistic regression analysis, we identified glucan-branching enzyme 1 (GBE1) as the most confident glycolytic gene implicated in PD. We validated the low expression of GBE1 in 1 - methyl - 4 - phenyl - 1,2,3,6 - tetrahydropyridine (MPTP)-induced PD animal models. Stereotaxic injection-mediated overexpression of GBE1 in striatal neurons improved motor dysfunction in these animal models. In vitro experiments demonstrated that GBE1 promotes the expression of lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB), enhances cellular glycolytic flux, and thereby increases the viability of PC12 cells under MPP+ treatment. Additionally, GBE1 alleviates mitochondrial dysfunction and restores oxidative phosphorylation in PD. In summary, by integrating machine learning and bioinformatics approaches, we identified GBE1, a glycolysis-related gene with significant implications for PD, elucidating its crucial role in glucose metabolic reprogramming and identifying potential therapeutic targets for modulating glucose metabolism in PD.
Collapse
Affiliation(s)
- Hongyan Chen
- Department of Neurology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai 201908, China.
| | - Hao Ding
- Department of Neurology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai 201908, China
| | - Dongya Huang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Shuo Wu
- Department of Neurology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai 201908, China.
| |
Collapse
|
11
|
Kim SJ, Lee K, Lee HJ, Kang DY, Kim YH. Maximum standardized uptake value-to-tumor size ratio in fluorodeoxyglucose F18 positron emission tomography/computed tomography: a simple prognostic parameter for non-small cell lung cancer. Diagn Interv Radiol 2025; 31:274-279. [PMID: 39354721 PMCID: PMC12057529 DOI: 10.4274/dir.2024.242837] [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: 05/08/2024] [Accepted: 08/24/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE By correcting the effect of tumor size on metabolic activity, the maximum standardized uptake value-to-tumor size (SUVmax:tumor size) ratio on fluorodeoxyglucose F18 positron emission tomography (18F-FDG PET)/computed tomography (CT) scans can be a prognostic parameter of non-small cell lung cancer (NSCLC). The current study evaluates the prognostic value of SUVmax:tumor size ratio on pretreatment 18F-FDG PET/CT scans in patients with NSCLC. Furthermore, the SUVmax:tumor size ratio is compared with other established PET parameters. METHODS This study included 108 patients with NSCLC who underwent pretreatment 18F-FDG PET/CT scans and curative lung surgery. The associations between the SUVmax:tumor size ratio and other conventional PET parameters were investigated. The recurrence-free survival according to the SUVmax:tumor size ratio was also analyzed. In addition, the SUVmax:tumor size ratio was compared according to postoperative pathologic findings. RESULTS In total, 72 (66.7%) of the 108 participants presented with adenocarcinoma (ADC). Nineteen (17.6%) patients experienced recurrence during a median follow-up period of 32.3 months. The median SUV max:tumor size ratio was 2.37 (1.23 for ADCs and 3.90 for other histologic types). The SUVmax:tumor size ratio was associated with SUVmax and mean SUV, as well as metabolic tumor volume and total lesion glycolysis. Patients with an SUVmax:tumor size ratio higher than the median had a worse recurrence outcome than those with an SUVmax:tumor size ratio lower than the median. Participants with ADC who presented with lymphovascular invasion had a higher SUVmax:tumor size ratio than those without. The presence of lymph node metastasis and advanced histologic grade were associated with a high SUVmax:tumor size ratio in patients with ADC. CONCLUSION The SUVmax:tumor size ratio on pretreatment 18F-FDG PET/CT scans was associated with aggressive tumor behavior and poor outcome in NSCLCs, particularly ADC. CLINICAL SIGNIFICANCE The SUVmax:tumor size ratio on pretreatment 18F-FDG PET/CT scans has a prognostic value in patients with NSCLCs, especially ADC.
Collapse
Affiliation(s)
- Soo Jeong Kim
- Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine , Department of Nuclear Medicine, Seoul, Republic of Korea
| | - Koeun Lee
- Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine , Department of Nuclear Medicine, Seoul, Republic of Korea
| | - Hyun Joo Lee
- Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine, Department of Pathology, Seoul, Republic of Korea
| | - Du-Young Kang
- Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine, Department of Thoracic and Cardiovascular Surgery, Seoul, Republic of Korea
| | - Young Hwan Kim
- Kangbuk Samsung Hospital Sungkyunkwan University School of Medicine , Department of Nuclear Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Schmidt M, Binder H, Schneider MR. The metabolic underpinnings of sebaceous lipogenesis. Commun Biol 2025; 8:670. [PMID: 40289206 PMCID: PMC12034822 DOI: 10.1038/s42003-025-08105-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Sebaceous glands synthesize and secrete sebum, a mélange of lipids and other cellular products that safeguards the mammalian integument. Differentiating sebocytes delaminate from the basal membrane and dislodge towards the gland's middle, where they eventually undergo a poorly understood death mode in which the whole cell becomes a secretion product (holocrine secretion). Supported by recent transcriptomics data, this review examines the idea that peripheral sebocytes have a remarkable ability to draw nutrients from the blood and become committed to unrestrainedly invest all available resources into synthetic processes for accomplishing sebum synthesis, thereby exploiting core metabolic fluxes as glycogen turnover, glutamine-directed anaplerosis, the pentose phosphate pathway and de novo lipogenesis. Finally, we propose that metabolic-driven processes are an important mechanistic component of holocrine secretion. A deeper understanding of these metabolic adaptations could indicate novel strategies for modulating sebum synthesis, a key pathogenic factor in acne and other skin diseases.
Collapse
Affiliation(s)
- Maria Schmidt
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
| | - Hans Binder
- Interdisciplinary Institute for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Armenian Bioinformatics Institute (ABI), Yerevan, Armenia
| | - Marlon R Schneider
- Institute of Veterinary Physiology, Veterinary Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
13
|
Paál Á, Dora D, Takács Á, Rivard C, Pickard SL, Hirsch FR, Roskó B, Kiraly P, Ferdinandy P, Varga ZV, Lohinai Z, Görbe A. Roles of Annexin A1 Expression in Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1407. [PMID: 40361334 PMCID: PMC12070913 DOI: 10.3390/cancers17091407] [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: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Small cell lung cancer (SCLC) is one of the malignancies with the worst prognosis, and there have been no major breakthroughs in its treatment for a long time. The majority of patients are diagnosed at the extensive stage, where the only option is chemotherapy, and even the addition of immune checkpoint inhibitors results in only modest benefits. The characterization of the molecular mechanisms behind therapy resistance has relevance in finding novel therapeutic approaches. Previous studies showed the possibility of annexin A1's (ANXA1) involvement in the immunosuppressive tumor microenvironment in SCLC, and there are studies showing the direct effects of ANXA1 modulation on cancer cell aggressiveness. METHODS We aimed to characterize the roles of ANXA1 expression using publicly available transcriptomic data, the RNA-seq-based predictive algorithms EPIC and ESTIMATE, and immunohistochemistry on patient samples. For the in vitro studies, we silenced ANXA1 expression with short hairpin RNA in three SCLC cell lines, measured the growth rate with the trypan blue exclusion assay, assessed the chemosensitivity to cisplatin and etoposide with the Presto BlueTM viability assay, and performed Western blots to assess changes in the levels of metabolic and mesenchymal markers and transcriptional drivers. RESULTS ANXA1-high tumors are associated with significantly increased immune infiltrates, stromality, and tumor-associated macrophages (TAMs). The ANXA1 protein is expressed on tumor cells and TAMs at the tissue level. ANXA1 silencing in H841 cells did not affect the growth rate; in SW1271 cells, shANXA1 cells grew significantly slower than shCTRL cells. Meanwhile, in H1048 cells, proliferation was significantly faster. Despite the different growth rates of the tested cell lines, ANXA1 silencing decreased the chemosensitivity to both cisplatin and etoposide in all three cell lines. Gene expression changes in mesenchymal markers, metabolic markers, dominant transcriptional drivers, and immune-relevant molecules were also characterized. CONCLUSIONS This is the first comprehensive characterization of ANXA1 in SCLC to reveal its role in the tumor's cell biology and the TME, aiming to boost further research in the field.
Collapse
Affiliation(s)
- Ágnes Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Ákos Takács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
| | - Christopher Rivard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Shivaun Lueke Pickard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Fred R. Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
- Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health System, New York, NY 10029, USA
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Peter Kiraly
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
14
|
Niphadkar S, Sreedharan S, Vengayil V, Laxman S. Protocol to quantitatively assess glycolysis and related carbon metabolic fluxes using stable isotope tracing in Crabtree-positive yeasts. STAR Protoc 2025; 6:103786. [PMID: 40266845 DOI: 10.1016/j.xpro.2025.103786] [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: 12/19/2024] [Revised: 03/14/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Crabtree-positive yeasts rapidly consume glucose via glycolysis, making it difficult to experimentally estimate their actual glycolytic rate or flux. We present a stable isotope labeling and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based protocol to quantitatively estimate glycolytic and related carbon metabolic fluxes using Saccharomyces cerevisiae. This approach defines time windows to capture glucose metabolic intermediate production before label saturation, enabling a comparison of glycolytic flux changes across different cells. This protocol provides a reliable, quantitative approach to study dynamic metabolic fluxes in these cells. For complete details on the use and execution of this protocol, please refer to Vengayil et al., 2024.1.
Collapse
Affiliation(s)
- Shreyas Niphadkar
- Institute for Stem Cell Science and Regenerative Medicine (BRIC inStem), GKVK Post Bellary Road, Bangalore 560065, India
| | - Sreesa Sreedharan
- Institute for Stem Cell Science and Regenerative Medicine (BRIC inStem), GKVK Post Bellary Road, Bangalore 560065, India; School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Vineeth Vengayil
- Institute for Stem Cell Science and Regenerative Medicine (BRIC inStem), GKVK Post Bellary Road, Bangalore 560065, India
| | - Sunil Laxman
- Institute for Stem Cell Science and Regenerative Medicine (BRIC inStem), GKVK Post Bellary Road, Bangalore 560065, India.
| |
Collapse
|
15
|
Li J, Liu H, Yang P, Zhu F, Shen F, Liang G. Identifying Aberrant 1CM-Related Pathways by Multi-Omics Analysis and Validating Tumor Inhibitory Effect of One-Carbon Donor Betaine in Gastric Cancer. Int J Mol Sci 2025; 26:3841. [PMID: 40332533 PMCID: PMC12027648 DOI: 10.3390/ijms26083841] [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: 03/09/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic reprogramming, a well-established hallmark of gastric carcinogenesis, has been implicated in driving tumor progression. Nevertheless, the precise mechanisms through which these metabolic alterations orchestrate gastric cancer (GC) pathogenesis remain incompletely elucidated. We conducted metabolomic analyses of plasma samples obtained from 334 patients with GC and healthy individuals to identify differential metabolites and metabolic pathways. Transcriptome sequencing was conducted on six pairs of tissues, and a joint analysis of the transcriptome and metabolome was performed. Single-cell sequencing data were acquired and co-analyzed with metabolomics to investigate metabolic abnormalities at the single-cell level. Finally, four representative metabolites selected using Random Forest analysis were subjected to cellular experiments to elucidate the mechanisms through which these metabolites exert their effects. Metabolomic analyses revealed that serine and glycine metabolism, glycolysis, and glutamate metabolism were significantly altered in GC, suggesting that one-carbon metabolism (1CM)-related pathways are aberrantly activated. A combined analysis of the transcriptome, single-cell transcriptome, and metabolomics indicated that pathways related to oxidative phosphorylation, nucleotide metabolism, and amino acid metabolism in epithelial cells were altered in GC. Cellular experiments demonstrated that the one-carbon donor metabolite betaine could inhibit the activity, invasion, and migration of GC cells while activating the phosphorylation of AMPKα. In conclusion, the 1CM-related pathway and the metabolite betaine play significant roles in GC, and the mechanisms through which the one-carbon donor betaine influences GC warrant further investigation.
Collapse
Affiliation(s)
- Jie Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.L.); (H.L.); (P.Y.)
| | - Huan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.L.); (H.L.); (P.Y.)
| | - Panpan Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.L.); (H.L.); (P.Y.)
| | - Feng Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Rd, Nanjing 210009, China; (F.Z.); (F.S.)
| | - Fei Shen
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Rd, Nanjing 210009, China; (F.Z.); (F.S.)
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.L.); (H.L.); (P.Y.)
| |
Collapse
|
16
|
Mussalo L, Afonin AM, Zavodna T, Krejcik Z, Honkova K, Fayad C, Shahbaz MA, Kalapudas J, Penttilä E, Löppönen H, Koivisto AM, Malm T, Topinka J, Jalava P, Lampinen R, Kanninen KM. Traffic-related ultrafine particles influence gene regulation in olfactory mucosa cells altering PI3K/AKT signaling. ENVIRONMENT INTERNATIONAL 2025; 199:109484. [PMID: 40273555 DOI: 10.1016/j.envint.2025.109484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/10/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Traffic-related ultrafine particles (UFPs) are an emerging health concern affecting the brain and increasing the risk of Alzheimer's disease (AD). PI3K/AKT signaling is known to contribute to neuronal survival and to be altered in AD. The nasal olfactory mucosa (OM) is a sensory tissue exposed directly to ambient air, and a starting point for olfactory neural circuits towards the brain. Evidence of air pollution-induced transcriptional regulation via microRNAs (miRNA) and DNA methylation (DNAmet) is accumulating and air pollutant-mediated disturbances in PI3K/AKT signaling have been reported. By utilizing a highly translational human-based in vitro model of OM, we aimed to investigate possible gene regulatory mechanisms in PI3K/AKT signaling induced by UFPs, and to compare the responses between cognitively healthy and individuals with AD. miRNA expression was analyzed using next-generation sequencing (NGS) and chip-based methylation analysis was performed to detect differentially methylated loci (DML). These data were combined with previously published transcriptomics analysis (mRNA) to construct an mRNA-miRNA-DNAmet-integrative network. Protein level changes were studied by immunoassays. We observed UFP-induced reductions in viability and increases in oxidative stress and DNA damage without eminent cell death. Integrative network analysis revealed multiple connections of miRNAs to differentially expressed genes in the PI3K/AKT pathway, and effects were most prominent in AD cells. Similarly, in AD cells DML were identified in transcription factor and apoptosis genes, downstream of PI3K/AKT signaling. Conclusively, traffic-related UFPs influence gene regulation of PI3K/AKT signaling to modulate OM cell survival, with existing AD pathology resulting in heightened vulnerability to UFP effects.
Collapse
Affiliation(s)
- Laura Mussalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Alexey M Afonin
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Tana Zavodna
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Zdenek Krejcik
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Katerina Honkova
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Claire Fayad
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Muhammad Ali Shahbaz
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Juho Kalapudas
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Elina Penttilä
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Heikki Löppönen
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anne M Koivisto
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210 Kuopio, Finland; Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, 70210 Kuopio, Finland; Department of Neurology and Geriatrics, Helsinki University Hospital and Neurosciences, Faculty of Medicine, University of Helsinki 00014 Helsinki, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Pasi Jalava
- Inhalation Toxicology Laboratory, Department of Environmental and Biological Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Riikka Lampinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Katja M Kanninen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland.
| |
Collapse
|
17
|
Ghanbarian M, Dolgova N, Vizeacoumar FS, Vizeacoumar FJ, Michel D, El-Aneed A, Dmitriev OY. Metabolic Effects of the Cancer Metastasis Modulator MEMO1. Metabolites 2025; 15:277. [PMID: 40278406 PMCID: PMC12029338 DOI: 10.3390/metabo15040277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Cancer cells often display altered energy metabolism. In particular, expression levels and activity of the tricarboxylic acid cycle (TCA cycle) enzymes may change in cancer, and dysregulation of the TCA cycle is a frequent hallmark of cancer cell metabolism. MEMO1, a modulator of cancer metastasis, has been shown to bind iron and regulate iron homeostasis in the cells. MEMO1 knockout changed mitochondrial morphology and iron content in breast cancer cells. Our previous genome-wide analysis of MEMO1 genetic interactions across multiple cancer cell lines revealed that gene sets involved in mitochondrial respiration and the TCA cycle are enriched among the gain-of-function interaction partners of MEMO1. Based on these findings, we measured the TCA cycle metabolite levels in breast cancer cells with varying levels of MEMO1 expression. Methods: ShRNA knockdown assay was performed to test essentiality of key TCA cycle enzymes. TCA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in MDA-MB-231 (high MEMO1), M67-2 (MEMO1 knockdown), and M67-9 (MEMO1 knockout) cells under iron-depleted, basal iron, and iron-supplemented conditions. Results:ACO2 and OGDH knockdowns inhibit cell proliferation, indicating an essential role of the TCA cycle in MDA-MB-231 metabolism. α-Ketoglutarate and citrate levels exhibited an inverse relationship with MEMO1 expression, increasing significantly in MEMO1 knockout cells regardless of iron availability. In contrast, fumarate, malate, and glutamate levels were elevated in MEMO1 knockout cells specifically under low iron conditions, suggesting an iron-dependent effect. Conclusions: Overall, our results indicate that MEMO1 plays a role in regulating the TCA in cancer cells in an iron-dependent manner.
Collapse
Affiliation(s)
- Marziyeh Ghanbarian
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| | - Natalia Dolgova
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| | - Frederick S. Vizeacoumar
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Franco J. Vizeacoumar
- Cancer Research Department, Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
- Division of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Deborah Michel
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anas El-Aneed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Oleg Y. Dmitriev
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.G.)
| |
Collapse
|
18
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [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: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
19
|
Zheng E, Gao W, Lu Q, Deng X, Xu S, Liu Z, Zeng L, Liu L, Zhang Q, Song H. Self-assembled metal-coordinated nanoparticles for synergistic energy metabolism inhibition and low-temperature photothermal therapy. Int J Pharm 2025; 674:125467. [PMID: 40089042 DOI: 10.1016/j.ijpharm.2025.125467] [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: 01/09/2025] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Photothermal therapy has been observed to upregulate the heat shock protein 70 (HSP 70) expression in tumor cells, consequently diminishing the anti-tumor efficacy of the treatment. The expression of HSP 70 is intricately linked to the adenosine triphosphate (ATP) levels within tumors, suggesting that modulating energy metabolism could potentially enhance the effectiveness of photothermal therapy. To address these challenges, ATO-QUE-Fe2+-PVP K30 nanoparticles (AQFP NPs) were synthesized through the coordinated self-assembly of the oxidative phosphorylation (OXPHOS) inhibitor atovaquone (ATO) and the glycolysis inhibitor quercetin (QUE) with ferrous ions (Fe2+) for synergetic energy depletion and low-temperature photothermal therapy (LTPTT). The synthesized AQFP NPs exhibited a small particle size and demonstrated high encapsulation efficiency of ATO and QUE. AQFP NPs could effectively downregulate the expression of HSP 70 by inhibiting the activity of mitochondrial complex Ⅲ and hexokinase Ⅱ (HK Ⅱ) to inhibiting suppress mitochondrial OXPHOS and glycolytic pathways in 4T1 cells, respectively. This inhibition resulted in a reduction of ATP levels within tumor cells, subsequently leading to decreased expression of HSP 70 and enhancing the therapeutic efficacy of LTPTT. Furthermore, AQFP NPs can remarkably inhibit the growth of tumors when subjected to laser irradiation. Furthermore, the analysis of blood biochemical indices and hematoxylin and eosin (H&E) staining of major organs suggested that AQFP NPs exhibit a preferable in vivo safety profile. In conclusion, the anti-tumor efficacy of LTPTT could be substantially enhanced by concurrently inhibiting OXPHOS and glycolysis, thereby offering an innovative therapeutic for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Enqin Zheng
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Wenhao Gao
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China; School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Qingyu Lu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Xiaoliang Deng
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Shiting Xu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China; School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhihong Liu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Lingjun Zeng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Linlin Liu
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Qian Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Hongtao Song
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China.
| |
Collapse
|
20
|
Icard P, Alifano M, Simula L. Citrate oscillations during cell cycle are a targetable vulnerability in cancer cells. Biochim Biophys Acta Rev Cancer 2025; 1880:189313. [PMID: 40216092 DOI: 10.1016/j.bbcan.2025.189313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
Cell cycle progression is timely interconnected with oscillations in cellular metabolism. Here, we first describe how these metabolic oscillations allow cycling cells to meet the bioenergetic needs specifically for each phase of the cell cycle. In parallel, we highlight how the cytosolic level of citrate is dynamically regulated during these different phases, being low in G1 phase, increasing in S phase, peaking in G2/M, and decreasing in mitosis. Of note, in cancer cells, a dysregulation of such citrate oscillation can support cell cycle progression by promoting a deregulated Warburg effect (aerobic glycolysis), activating oncogenic signaling pathways (such as PI3K/AKT), and promoting acetyl-CoA production via alternative routes, such as overconsumption of acetate. Then, we review how administration of sodium citrate (at high doses) arrests the cell cycle in G0/G1 or G2/M, inhibits glycolysis and PI3K/AKT, induces apoptosis, and significantly reduces tumor growth in various in vivo models. Last, we reason on the possibility to implement citrate administration to reinforce the effectiveness of cell cycle inhibitors to better cure cancer.
Collapse
Affiliation(s)
- Philippe Icard
- Université de Normandie, UNICAEN, Inserm U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Caen, France; Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Descartes, Paris, France.
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Descartes, Paris, France; Inserm U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Luca Simula
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Paris 75014, France
| |
Collapse
|
21
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
22
|
Markov N, Sabirova S, Sharapova G, Gomzikova M, Brichkina A, Barlev NA, Egger M, Rizvanov A, Simon HU. Mitochondrial, metabolic and bioenergetic adaptations drive plasticity of colorectal cancer cells and shape their chemosensitivity. Cell Death Dis 2025; 16:253. [PMID: 40185729 PMCID: PMC11971274 DOI: 10.1038/s41419-025-07596-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
The extent of mitochondrial heterogeneity and the presence of mitochondrial archetypes in cancer remain unknown. Mitochondria play a central role in the metabolic reprogramming that occurs in cancer cells. This process adjusts the activity of metabolic pathways to support growth, proliferation, and survival of cancer cells. Using a panel of colorectal cancer (CRC) cell lines, we revealed extensive differences in their mitochondrial composition, suggesting functional specialisation of these organelles. We differentiated bioenergetic and mitochondrial phenotypes, which point to different strategies used by CRC cells to maintain their sustainability. Moreover, the efficacy of various treatments targeting metabolic pathways was dependent on the respiration and glycolysis levels of cancer cells. Furthermore, we identified metabolites associated with both bioenergetic profiles and cell responses to treatments. The levels of these molecules can be used to predict the therapeutic efficacy of anti-cancer drugs and identify metabolic vulnerabilities of CRC. Our study indicates that the efficacy of CRC therapies is closely linked to mitochondrial status and cellular bioenergetics.
Collapse
Affiliation(s)
- Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Sirina Sabirova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Gulnaz Sharapova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Nick A Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Marcel Egger
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany.
| |
Collapse
|
23
|
Majhi S, Roy P, Jo M, Liu J, Hurto R, Freddolino L, Marsh ENG. Viperin expression leads to downregulation of mitochondrial genes through misincorporation of ddhCTP by mitochondrial RNA polymerase. J Biol Chem 2025; 301:108359. [PMID: 40015636 PMCID: PMC11982959 DOI: 10.1016/j.jbc.2025.108359] [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: 12/13/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Increasing lines of evidence link the expression of the interferon-stimulated gene RSAD2, encoding the antiviral enzyme, viperin, to autoimmune disease. Autoimmune diseases are characterized by chronic overproduction of cytokines such as interferons that upregulate the inflammatory response. Immune cells exposed to interferon selectively downregulate transcription of the mitochondrially encoded components of the oxidative phosphorylation system, which leads to mitochondria becoming dysfunctional and impairing their ability to produce ATP. But the mechanism by which downregulation occurs has remained unknown. Here we show that 3'-deoxy-3',4'-didehydrocytidine triphosphate (ddhCTP) which is synthesized by viperin suppresses mitochondrial transcription by causing premature chain termination when misincorporated by the mitochondrial RNA polymerase (POLRMT). We show that viperin expression in human cell lines downregulates mitochondrially encoded gene expression. A similar effect is observed across multiple cell lines when cells are exposed to ddhC, the precursor to ddhCTP. The pattern of gene downregulation fits well with a simple, quantitative model describing chain-termination. In vitro measurements with purified POLRMT demonstrate that ddhCTP competes effectively with CTP, leading to its misincorporation into RNA. These findings reveal a new molecular mechanism for mitochondrial transcriptional regulation that explains the reduction in mitochondrially-encoded transcript levels in response to chronic interferon stimulation, characteristic of inflammatory diseases.
Collapse
Affiliation(s)
- Srijoni Majhi
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Pronay Roy
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Minshik Jo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiying Liu
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Rebecca Hurto
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lydia Freddolino
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - E Neil G Marsh
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA; Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
24
|
Atamna B, Rozental A, Haj Yahia M, Itchaki G, Gurion R, Yeshurun M, Raanani P, Wolach O. Tumor-Associated Lactic Acidosis and Early Death in Patients With Lymphoma. Cancer Med 2025; 14:e70824. [PMID: 40152524 PMCID: PMC11951173 DOI: 10.1002/cam4.70824] [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: 11/14/2024] [Revised: 12/30/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Cancer is characterized by accelerated glycolysis with enhanced glucose uptake and lactate production, a phenomenon termed Warburg effect (WE). We studied the incidence and clinical impact of Warburg-driven lactic acidosis in lymphoma. METHODS Patients admitted with newly diagnosed or relapsed/refractory lymphoma and documented lactate levels during the first week of admission were included. Patients with lactatemia were classified as secondary (with a recognizable cause for elevated lactate) or none (WE group). RESULTS WE and secondary lactatemia were documented in 58 and 44 patients (15% and 12% of evaluable patients, respectively). Both WE and secondary lactatemia were associated with poor short-term survival. WE at presentation correlated with tumor burden, with most patients having aggressive disease, advanced stage, and extranodal involvement. WE was associated with high rates of early death (26% and 43% at 30- and 60-days, respectively). Higher lactate levels correlated with worse survival. Earlier initiation of chemotherapy was associated with a (nonsignificant) trend toward better outcomes, whereas steroid and/or thiamine therapy did not alter patient outcomes. Glucose administration was associated with worse survival. CONCLUSION WE-driven lactatemia is associated with high tumor burden and increased short-term mortality in lymphoma. Prompt initiation of anti-lymphoma therapy may improve outcomes.
Collapse
Affiliation(s)
- Bahaa Atamna
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Alon Rozental
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Mohammad Haj Yahia
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
| | - Gilad Itchaki
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
- Institute of HematologyMeir Medical CenterKfar‐SabaIsrael
| | - Ronit Gurion
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Moshe Yeshurun
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Ofir Wolach
- Institute of Hematology, Davidoff Cancer CenterRabin Medical CenterPetah‐TikvaIsrael
- Faculty of Medical & Health SciencesTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
25
|
Harada Y. Manipulating mannose metabolism as a potential anticancer strategy. FEBS J 2025; 292:1505-1519. [PMID: 39128015 DOI: 10.1111/febs.17230] [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: 03/06/2024] [Revised: 05/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
Cancer cells acquire metabolic advantages over their normal counterparts regarding the use of nutrients for sustained cell proliferation and cell survival in the tumor microenvironment. Notable among the metabolic traits in cancer cells is the Warburg effect, which is a reprogrammed form of glycolysis that favors the rapid generation of ATP from glucose and the production of biological macromolecules by diverting glucose into various metabolic intermediates. Meanwhile, mannose, which is the C-2 epimer of glucose, has the ability to dampen the Warburg effect, resulting in slow-cycling cancer cells that are highly susceptible to chemotherapy. This anticancer effect of mannose appears when its catabolism is compromised in cancer cells. Moreover, de novo synthesis of mannose within cancer cells has also been identified as a potential target for enhancing chemosensitivity through targeting glycosylation pathways. The underlying mechanisms by which alterations in mannose metabolism induce cancer cell vulnerability are just beginning to emerge. This review summarizes the current state of our knowledge of mannose metabolism and provides insights into its manipulation as a potential anticancer strategy.
Collapse
Affiliation(s)
- Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Research Institute, Osaka International Cancer Institute, Japan
| |
Collapse
|
26
|
Chi XK, Zhang HR, Gao JJ, Su J, Du YZ, Xu XL. Polydopamine-based Nanoadjuvants Promote a Positive Feedback Loop for Cancer Immunotherapy via Overcoming Photothermally Boosted T Cell Exhaustion. Biomater Res 2025; 29:0166. [PMID: 40110052 PMCID: PMC11922554 DOI: 10.34133/bmr.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Immunogenic cell death, triggered by photothermal therapy or specific chemotherapy, strives to establish a positive feedback loop in cancer immunotherapy. This loop is characterized by the rapid release of antigens and adenosine triphosphate (ATP), ultimately leading to accelerated T cell infiltration. However, this loop is hindered by T cell exhaustion caused by adenosine originating from ATP and glucose deprivation in the immunosuppressive microenvironment. To overcome this challenge, we developed a pH-low insertion peptide-functionalized mesoporous-polydopamine-based nanoadjuvant that incorporates adenosine deaminase and doxorubicin (termed as PPMAD). PPMAD aimed to overcome T cell exhaustion by reducing adenosine consumption and providing an alternative carbon source for CD8+ T cell function during glucose starvation. First, PPMAD triggered the burst release of antigens and ATP through photothermal therapy and doxorubicin-induced immunogenic cell death, culminating in the expedited infiltration of T cells. Second, adenosine deaminase depleted adenosine, reducing immunosuppressive agents and generating abundant inosine, which served as an alternative carbon source for CD8+ T cells. By implementing this "reducing suppression and broadening sources" strategy, we successfully overcome T cell exhaustion, greatly enhancing the effectiveness of cancer immunotherapy both in vitro and in vivo. Our findings highlighted the positive feedback loop between on-demand photothermal therapy, chemotherapy immunotherapy, and achieving complete tumor response.
Collapse
Affiliation(s)
- Xiao-Kai Chi
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Hai-Rui Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jing-Jing Gao
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jin Su
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| |
Collapse
|
27
|
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
|
28
|
Nourbakhsh ST, Mirzaei SA, Mohamadhashem F, Naghizadeh MM, Razavi AN, Mansoori Y, Daraei A, Mohamadhashem F. Pathological expression of mitochondrial genome-derived circRNA SCAR/mc-COX2 and its ceRNA network in colorectal cancer: implications for clinical significance. BMC Cancer 2025; 25:466. [PMID: 40082804 PMCID: PMC11907809 DOI: 10.1186/s12885-025-13886-7] [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: 01/09/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Mitochondrial-encoded circular RNAs (mecciRNAs) are a newly discovered class of mitochondrial-encoded non-coding RNAs (mt-ncRNAs) that play important biological roles in the cell. This study aimed to examine the expression profile of SCAR/mc-COX2 (has_circ_0089762) in colorectal cancer (CRC) and its relationship with clinicopathological variables. Furthermore, to better understand SCAR/mc-COX2's functional role in CRC, we constructed a competing endogenous RNA (ceRNA) network. METHODS Quantitative real-time PCR (qRT-PCR) was employed to analyze the expression levels of SCAR/mc-COX2 in 40 pairs of CRC samples, consisting of 40 tumor samples and 40 adjacent non-tumoral samples from patients. The ceRNA regulatory network was constructed using online bioinformatics tools. Furthermore, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) enrichment analysis were conducted using the Enrichr database. RESULTS The results demonstrated a significant decrease in SCAR/mc-COX2 expression in tumor tissues compared to adjacent non-tumoral tissues (p-value<0.05). In another finding, a significant relationship was observed between pathological T staging and the expression status of SCAR/mc-COX2 (p-value=0.02). Additionally, the Receiver Operating Characteristic (ROC) curve analysis revealed that SCAR/mc-COX2 had an area under the curve (AUC) of 0.77, with 80% sensitivity and 75% specificity. Finally, a ceRNA regulatory network including SCAR/mc-COX2, 5 miRNA, and 9 mRNAs was found. CONCLUSION These findings suggest that SCAR/mc-COX2 may act as a tumor suppressor in CRC, and its dysregulation could play a crucial role in the pathophysiology of this cancer. The significant association with pathological T staging and its robust diagnostic performance (AUC = 0.77, sensitivity = 80%, specificity = 75%) highlight its potential as a novel biomarker for CRC detection and prognosis. Further functional studies are required to elucidate its precise role in CRC tumorigenesis and clinical applicability.
Collapse
Affiliation(s)
- Seyed Taha Nourbakhsh
- Department of Medical Genetics, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Mohamadhashem
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amir Nader Razavi
- Iran National Tumor Bank, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Faezeh Mohamadhashem
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
29
|
Wang FX, Mu G, Yu ZH, Shi ZA, Li XX, Fan X, Chen Y, Zhou J. Lactylation: a promising therapeutic target in ischemia-reperfusion injury management. Cell Death Discov 2025; 11:100. [PMID: 40082399 PMCID: PMC11906755 DOI: 10.1038/s41420-025-02381-4] [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: 03/02/2024] [Revised: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Ischemia-reperfusion injury (IRI) is a critical condition that poses a significant threat to patient safety. The production of lactate increases during the process of IRI, and lactate serves as a crucial indicator for assessing the severity of such injury. Lactylation, a newly discovered post-translational modification in 2019, is induced by lactic acid and predominantly occurs on lysine residues of histone or nonhistone proteins. Extensive studies have demonstrated the pivotal role of lactylation in the pathogenesis and progression of various diseases, including melanoma, myocardial infarction, hepatocellular carcinoma, Alzheimer's disease, and nonalcoholic fatty liver disease. Additionally, a marked correlation between lactylation and inflammation has been observed. This article provides a comprehensive review of the mechanism underlying lactylation in IRI to establish a theoretical foundation for better understanding the interplay between lactylation and IRI.
Collapse
Affiliation(s)
- Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Guo Mu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People's Hospital, Zigong, Sichuan, China
| | - Zu-An Shi
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Xue-Xin Li
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Fan
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China.
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
30
|
Li J, Li S, Sun Q, Li L, Zhang Y, Hua Z. H3K18 lactylation-mediated nucleotide-binding oligomerization domain-2 (NOD2) expression promotes bilirubin-induced pyroptosis of astrocytes. J Neuroinflammation 2025; 22:76. [PMID: 40075479 PMCID: PMC11905654 DOI: 10.1186/s12974-025-03399-2] [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: 08/14/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Histone lactylation, a newly glycosis-related histone modification, plays a crucial role in the regulation of gene expression in various immune cells. However, the role of histone lactylation in astrocytes remains unclear. Here, this study showed that the H3K18 lactylation (H3K18la) levels were upregulated in primary astrocytes under unconjugated bilirubin (UCB) stimulation and hippocampus of bilirubin encephalopathy (BE) rats. Inhibition of glycolysis decreased H3K18la and attenuated pyroptosis both in vitro and in vivo. CUT& Tag and RNA-seq results revealed that H3K18la was enriched at the promoter of nucleotide-binding oligomerization domain 2 (NOD2) and promoted its transcription. Moreover, NOD2 boosted the activation of downstream mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signaling pathways, which exacerbated the neuroinflammation of BE. Collectively, this study provides a novel understanding of epigenetic regulation in astrocytes, and interruption of the H3K18la/NOD2 axis may represent a novel therapeutic strategy for treating bilirubin encephalopathy.
Collapse
Affiliation(s)
- Jing Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Siyu Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Qian Sun
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Ling Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Yan Zhang
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Ziyu Hua
- Department of Neonatology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| |
Collapse
|
31
|
Yao L, Gu C, Ge R, Zhang X, Meng X, Wang L, Peng D, Li G. Acetylated Dendrobium huoshanense polysaccharide: a novel inducer of apoptosis in colon cancer cells via Fas-FasL pathway activation and metabolic reprogramming. Front Oncol 2025; 15:1529868. [PMID: 40104499 PMCID: PMC11913854 DOI: 10.3389/fonc.2025.1529868] [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: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction Not all polysaccharides function as antitumor drugs, nor do they universally possess the same advantages regarding safety and biocompatibility. Those polysaccharides that are effective antitumor agents typically demonstrate superior safety profiles and biocompatibility compared to synthetic anticancer drugs, which can exhibit high toxicity and harmful side effects. Dendrobium huoshanense polysaccharide (DHP) has been recognized for its potential bioactive properties, particularly in anti-tumor treatment. This study investigates the effects of DHP on the proliferation and apoptosis of HCT116 colon cancer cells. Methods DHP was extracted according to previously published experimental methods. The inhibitory effects of DHP were evaluated using IEC6, Caco-2, and HCT116 cell lines, with changes in cell morphology observed via transmission electron microscopy. After establishing the conditions for DHP administration, flow cytometry was employed to assess its effects on apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential of HCT116 cells. Additionally, immunoprecipitation, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and biomarker detection were utilized to investigate the mechanisms underlying DHP's inhibition of HCT116 cells and its impact on metabolic reprogramming. Results In the present study, we observed that DHP treatment at 600 μg/ml for 24 h reduced HCT116 cell viability to 54.87%. In contrast, the inhibitory effect of DHP on the viability of IEC6 and Caco-2 cells was relatively mild. The specific mechanism involves DHP activating the mitochondrial apoptotic pathway leading to the downregulation of key metabolic intermediates and enzymes such as uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) and ST6Gal-I. By inhibiting ST6Gal-I activity, DHP activates the Fas/FasL signaling pathway. Additionally, DHP-induced ROS production effectively triggers apoptosis in HCT116 cells. Conclusion Our study demonstrates that DHP effectively inhibits the proliferation and induces apoptosis in HCT116 colon cancer cells through the activation of the Fas-FasL signaling pathway and metabolic reprogramming. The selective inhibitory effect of DHP on HCT116 cells, the activation of both death receptor and mitochondrial apoptotic pathways, and the modulation of metabolic reprogramming provide novel insights into the potential therapeutic strategies for colon cancer.
Collapse
Affiliation(s)
- Liang Yao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Chen Gu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Ruipeng Ge
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
| | - Xiaoqian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Xinqian Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
- Ministry of Education (MOE)-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory for Research and Development of Research and Development of Chinese Medicine, Hefei, China
| | - Guozhuan Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Academy of Chinese Medicine, Hefei, China
| |
Collapse
|
32
|
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
|
33
|
Zhang Y, Tang J, Jiang C, Yi H, Guang S, Yin G, Wang M. Metabolic reprogramming in cancer and senescence. MedComm (Beijing) 2025; 6:e70055. [PMID: 40046406 PMCID: PMC11879902 DOI: 10.1002/mco2.70055] [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: 07/06/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 04/01/2025] Open
Abstract
The rising trend in global cancer incidence has caused widespread concern, one of the main reasons being the aging of the global population. Statistical data show that cancer incidence and mortality rates show a clear upward trend with age. Although there is a commonality between dysregulated nutrient sensing, which is one of the main features of aging, and metabolic reprogramming of tumor cells, the specific regulatory relationship is not clear. This manuscript intends to comprehensively analyze the relationship between senescence and tumor metabolic reprogramming; as well as reveal the impact of key factors leading to cellular senescence on tumorigenesis. In addition, this review summarizes the current intervention strategies targeting nutrient sensing pathways, as well as the clinical cases of treating tumors targeting the characteristics of senescence with the existing nanodelivery research strategies. Finally, it also suggests sensible dietary habits for those who wish to combat aging. In conclusion, this review attempts to sort out the link between aging and metabolism and provide new ideas for cancer treatment.
Collapse
Affiliation(s)
- Yuzhu Zhang
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Jiaxi Tang
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Can Jiang
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Hanxi Yi
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Shu Guang
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Gang Yin
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Maonan Wang
- Department of PathologyXiangya HospitalSchool of Basic Medical SciencesCentral South UniversityChangshaChina
| |
Collapse
|
34
|
Li X, Liu B, Wang S, Dong Q, Li J. EDNRB negatively regulates glycolysis to exhibit anti-tumor functions in prostate cancer by cGMP/PKG pathway. Mol Cell Endocrinol 2025; 598:112459. [PMID: 39788311 DOI: 10.1016/j.mce.2025.112459] [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: 10/22/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer in men and the leading cause of cancer-related mortality. Recent studies have highlighted the pivotal role of glycolysis in tumor progression. This study aimed to investigate the involvement of the EDNRB gene and its ligand endothelin 3 (EDN3) in glycolysis in PCa and to elucidate its underlying molecular mechanism. Quantitative reverse transcription PCR (RT-qPCR) and methylation-specific PCR (MSP) were used to probe EDNRB expression and methylation in PCa tissues. Cell proliferation and glycolysis in PCa cells were evaluated using Cell Counting Kit-8 (CCK-8), EDU staining, Seahorse assay, and biochemical kits to analyze the effects of EDN3/EDNRB. The underlying molecular mechanism was further explored through Western blotting. The in vivo effect of EDNRB on tumor growth was examined using a xenograft tumor model. Our findings revealed that EDNRB was hypermethylated and downregulated in PCa tissues and cell lines. Overexpression of EDNRB or EDN3 led to reduced cell proliferation and downregulation of glycolytic markers. EDNRB also decreased the extracellular acidification rate (ECAR) baseline and increased the oxygen consumption rate (OCR) baseline, indicating a shift away from glycolysis. Additionally, the anticancer effects of EDNRB or EDN3 was reversed upon inhibition of the cGMP/PKG pathway. In vivo, enhanced EDNRB expression significantly suppressed tumor growth. Therefore, EDNRB or EDN3 possess anticancer potential in PCa, primarily through the regulation of glycolysis via the cGMP/PKG pathway.
Collapse
Affiliation(s)
- Xun Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Bide Liu
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Shuheng Wang
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Qiang Dong
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Jiuzhi Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China.
| |
Collapse
|
35
|
Tanaka AR, Murakami C, Yamamoto H. Methylmalonic acid at the serum level in the elderly contributes to cell growth via mitochondrial dysfunction in colorectal cancer cell spheroids. Biochem Biophys Rep 2025; 41:101909. [PMID: 39886070 PMCID: PMC11780164 DOI: 10.1016/j.bbrep.2024.101909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Methylmalonic acid (MMA) is a small molecule produced during the metabolism of propionate and branched-chain amino acids. Recently, it has been reported that the blood concentration of MMA increases with age and promotes lung cancer metastasis. However, little is known regarding its effects on cancers other than lung cancer. In the present study, we examined the effects of MMA on colorectal cancer cell spheroids. We found that MMA promoted the proliferation of colorectal cancer spheroids at physiological concentrations that can be exhibited by the elderly and induced mitochondrial reactive oxygen species generation, which in turn affected the promotion of cell growth. MMA treatment also induces a metabolic shift in the glycolytic system. These results suggest that MMA may promote cancer cell proliferation by decreasing mitochondrial function, inducing a metabolic shift, and provide new insights into the effects of aging on cancer.
Collapse
Affiliation(s)
- Arowu R. Tanaka
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami Ward, Hiroshima City, Hiroshima, 731-0153, Japan
| | - Chiho Murakami
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami Ward, Hiroshima City, Hiroshima, 731-0153, Japan
| | - Hideya Yamamoto
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami Ward, Hiroshima City, Hiroshima, 731-0153, Japan
| |
Collapse
|
36
|
Yao M, Zhang X, Wu T, Feng T, Bian X, Abudurexiti M, Wang W, Shi G, Wei GH, Zhang Q, Li X, Feng G, Du L, Wang J. Lon protease 1-mediated metabolic reprogramming promotes the progression of prostate cancer. Cell Death Dis 2025; 16:116. [PMID: 39971909 PMCID: PMC11840119 DOI: 10.1038/s41419-025-07449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/30/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
Lon protease 1 (LONP1) is an ATP-dependent protease located in the mitochondrial matrix and plays a crucial role in regulating mitochondrial proteostasis, metabolism, and cellular stress responses et al. Aberrant LONP1 expression has been found in the progression of various tumors; however, the role and molecular mechanisms of LONP1 in prostate cancer (PCa) remain poorly understood. Here we show that overexpression of LONP1 is closely related to adverse clinic pathological features and poor prognosis in PCa patients. Mechanistically, the findings reveal that LONP1 is implicated in modulating the metabolic switch from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, thereby promoting tumor proliferation, invasion, and metastasis both in vitro and in vivo. Meanwhile, we prove that LONP1 as a protease directly targets mitochondrial pyruvate carrier 1 (MPC1), a key metabolic protein in the process of glycolysis, and enhances its degradation, which in turn suppresses tricarboxylic acid (TCA) cycle and ultimately promotes the progression of PCa. Furthermore, using PCa in cancer-prone mice homozygous for a prostate-targeted conditional Pten knockout and Lonp1 knockin, we integrate transcriptomic and proteomic analyses of prostate tumors, upon which reveals that Lonp1 overexpression results in a significant downregulation of NADH: ubiquinone oxidoreductase activity, consequently impeding the electron transfer process and mitochondrial ATP synthesis, associated with metastasis of PCa. Collectively, our results highlight that metabolic reprogramming induced by LONP1 in PCa is closely coupled with disease progression, suggesting that targeting the LONP1-mediated cascade in the mitochondrial may provide therapeutic potential for PCa disease.
Collapse
Affiliation(s)
- Mengfei Yao
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Nuclear Medicine, Zhongshan Hospital affiliated Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xingming Zhang
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tianqi Wu
- Radiation Oncology Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Tao Feng
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaojie Bian
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mierxiati Abudurexiti
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Urology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Wenfeng Wang
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gong-Hong Wei
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200025, China
| | - Qin Zhang
- Disease Networks Research Unit, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014, Oulu, Finland
| | - Xiangyun Li
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gang Feng
- Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| | - Leilei Du
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jianhua Wang
- Cancer Institute, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| |
Collapse
|
37
|
Lu W, Wen J. Metabolic reprogramming and astrocytes polarization following ischemic stroke. Free Radic Biol Med 2025; 228:197-206. [PMID: 39756488 DOI: 10.1016/j.freeradbiomed.2025.01.002] [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: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Astrocytes are critical for maintaining neuronal activity. Activation of astrocytes, occurs within minutes from ischemic stroke onset due to ischemic causes and subsequent inflammatory damage. Activated astrocytes, also known as reactive astrocytes, are divided into two different phenotypes: A1 (pro-inflammatory) and A2 (anti-inflammatory) astrocytes. A2 astrocytes support neuronal survival and promote tissue healing, while A1 astrocytes have neurotoxic effects. Thus, polarization of reactive astrocyte into A1 or A2 genotype is closely correlated with the development of cerebral ischemia/reperfusion (I/R) injury. Metabolic reprogramming is a process that various metabolic pathways upregulate in cells to balance energy, alter their phenotype, and produce building-block requirements. A1 and A2 astrocytes display different metabolic reprogramming, such as glycolysis, glutamate uptake, and glycogenolysis. Accumulating evidence suggested that manipulation of energy metabolism homeostasis can induce astrocytes to switch from A1 to A2 phenotype. This review disucss the potential factors in affecting astrocytic polarization, emphasizes metabolic reprogramming in reactive astrocytes within the pathophysiological context of cerebral I/R, and explores the relationship between metabolic reprogramming and astrocytic polarization. Importantly, we reveal that regulating metabolic reprogramming in reactive astrocytes may be a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
38
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 PMCID: PMC11814237 DOI: 10.1038/s41392-024-02082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
39
|
Xiong Z, Huang Y, Cao S, Huang X, Zhang H. A new strategy for the treatment of advanced ovarian cancer: utilizing nanotechnology to regulate the tumor microenvironment. Front Immunol 2025; 16:1542326. [PMID: 40013141 PMCID: PMC11860879 DOI: 10.3389/fimmu.2025.1542326] [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: 12/09/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
Advanced ovarian cancer (AOC) is prone to recurrence, which can be attributed to drug resistance. Drug resistance may be related to the tumor microenvironment (TME), including the immune and non-immune TME. In the immune TME, the immune effector cells such as dendritic cells (DCs), M1-like tumor-associated macrophages (M1-TAMs), and T cells are inhibited. In contrast, immunosuppressive cells such as M2-like tumor-associated macrophages (M2-TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs) are activated. These changes make it difficult to produce immune effects and affect the efficacy of chemo-immunotherapy. In the non-immune TME, mechanisms such as apoptosis inhibition, DNA damage response (DDR), and epithelial-mesenchymal transition (EMT) can promote tumor growth, metastasis, and drug resistance. Despite the challenges posed by the TME in the treatment of AOC, the unique biological advantages of nanoparticles (NPs) make it possible to regulate the TME. NPs can stimulate the immune responses of M1-TAMs, DCs, and T cells while reducing the infiltration of immune suppressive cells such as M2-TAMs and Tregs, thereby regulating the AOC immune TME. In addition, NPs can regulate the non-immune TME by reducing apoptosis in AOC cells, inhibiting homologous recombination (HR) repair, reversing EMT, and achieving the effect of reversing drug resistance. In summary, the application of NPs provides some new venues for clinical treatment in AOC.
Collapse
Affiliation(s)
- Zixuan Xiong
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yichun Huang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shulong Cao
- Department of Pathology, Songzi People’s Hospital, Jingzhou, China
| | - Xuqun Huang
- Department of Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
40
|
Huang Z, Tian K, Xue Y, Luo F. A promising role of noble metal NPs@MOFs in chondrosarcoma management. NANOSCALE 2025; 17:2961-2984. [PMID: 39718125 DOI: 10.1039/d4nr03878a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Chondrosarcoma, a challenging and malignant neoplasm originating from cartilage cells, poses significant diagnostic and therapeutic hurdles due to its resistance to conventional treatments and the complexity of its diagnosis. Noble metal nanoparticle-embedded metal-organic frameworks (NPs@MOFs) stand out as a novel approach for the diagnosis and treatment of chondrosarcoma. This review delves into the properties and applications of NPs@MOFs, focusing on their classification by noble metal type and their role in enhancing photothermal therapy (PTT), photodynamic therapy (PDT), targeted drug delivery and chondrosarcoma diagnosis. Despite promising in vitro and in vivo results, challenges such as understanding the mechanisms of action and clinical translation remain, and the therapeutic effect of PTT and PDT on deep chondrosarcoma seems unsatisfactory. Future exploration, such as combined therapy and multiple MOF therapy, could unlock the full potential of noble metal NPs@MOFs in revolutionizing chondrosarcoma management, offering insights into the prospect of these materials in chondrosarcoma management.
Collapse
Affiliation(s)
- Ziheng Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Keyue Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yiyuan Xue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Department of General Dentistry, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China.
| |
Collapse
|
41
|
Wang X, Cheng L, Liu A, Liu L, Gong L, Shen G. Metabolomics approach reveals key plasma biomarkers in multiple myeloma for diagnosis, staging, and prognosis. J Transl Med 2025; 23:163. [PMID: 39915820 PMCID: PMC11800462 DOI: 10.1186/s12967-024-05848-7] [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: 07/26/2024] [Accepted: 10/30/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) is the most aggressive and prevalent primary malignant tumor within the blood system, and can be classified into grades RISS-I, II, and III. High-grade tumors are associated with decreased survival rates and increased recurrence rates. To better understand metabolic disorders and expand the potential targets for MM, we conducted large-scale untargeted metabolomics on plasma samples from MM patients and healthy controls (HC). METHODS Our study included 33 HC, 38 newly diagnosed MM patients (NDMM) categorized into three RISS grades (grade I: n = 5; grade II: n = 19; grade III: n = 8), and 92 MM patients post-targeted therapy with bortezomib-based regimens. Simultaneously, MM cell lines were employed for validation studies. Metabolites were analyzed and identified using ultra high liquid chromatography coupled with Q Orbitrap mass spectrometry (UPLC-HRMS), followed by verification through a self-built database. RESULTS Compared with HC participants, a total of 70 metabolites were identified as undergoing significant changes in NDMM. These metabolites were significantly enriched in citrate cycle, choline metabolism, glycerophospholipid metabolism, and sphingolipid metabolism, etc. Notably, a panel of circulating plasma metabolite biomarkers, including lactic acid and leucine, has emerged not only as diagnostic indicators but also as valuable tools for tumor surveillance, aiding in the assessment of disease stage and prognostic evaluation. Moreover, 14 differential metabolites were identified in both MM cell lines and MM patients. Among these, intracellular levels of lactate and leucine significantly decreased in vitro, aligning with the plasma results. CONCLUSION Our findings on key metabolites and metabolic pathways provide novel insights into the exploration of diagnostic and therapeutic targets for MM. A prospective study is essential to validate these discoveries for future MM patient care.
Collapse
Affiliation(s)
- Xiaoxue Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Longhao Cheng
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China
| | - Aijun Liu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Lihong Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China
| | - Lili Gong
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Capital Medical University, No. 2 YingHua Road, Beijing, 100029, China.
| | - Guolin Shen
- Institute of Chemicals Safety, Chinese Academy of Inspection and Quarantine, No. 11 Rong Hua Middle Road, Economic-Technological Development Area, Beijing, 100176, China.
| |
Collapse
|
42
|
Zhou Z, Li Q, Huo R. SUCLG1 promotes aerobic respiration and progression in plexiform neurofibroma. Int J Oncol 2025; 66:10. [PMID: 39749698 PMCID: PMC11753773 DOI: 10.3892/ijo.2024.5716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Plexiform neurofibromas (PNFs) are benign tumors that affect 20‑50% of patients with type I neurofibromatosis (NF1). PNF carries a risk of malignancy. There is no effective cure for PNF. Its onset may be associated with genetic and metabolic abnormalities, but the exact mechanisms remain unclear. Succinate‑CoA ligase GDP/ADP‑Forming Subunit α(SUCLG1), a catalytic enzyme in the tricarboxylic acid cycle, is highly expressed in PNF. The present study aimed to explore the role of SUCLG1 in function and metabolism of PNF cells. SUCLG1 expression was verified using western blotting and immunofluorescence. After inducing SUCLG1 knockdown and overexpression, functional changes in PNF cells were assessed, as well as effects of SUCLG1 on cell respiration and glucose metabolism. Quantitative PCR, WB, electron microscopy and Flow cytometry demonstrated that SUCLG1 enhanced mitochondrial quality and promoted mitochondrial fusion, thereby driving proliferation and migration of tumor cells, inhibiting apoptosis and altering the cell cycle. A Seahorse assay showed that elevated SUCLG1 expression enhanced cell aerobic respiration without affecting the glycolytic process. This suggests that SUCLG1 upregulation in PNF does not trigger the Warburg effect associated with malignant tumors. This study also demonstrated the positive regulation of cellular function by promoting the expression level of the SLC25A1 gene when SUCLG1 expression was elevated. In conclusion, SUCLG1 altered the mechanism of mitochondrial quality control to enhance cell aerobic respiration, thereby driving the pathogenesis of PNF. Thus, SUCLG1 can serve as a potential target in future therapeutic strategies.
Collapse
Affiliation(s)
- Zifu Zhou
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Ran Huo
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| |
Collapse
|
43
|
Peter A, Berneman ZN, Cools N. Cellular respiration in dendritic cells: Exploring oxygen-dependent pathways for potential therapeutic interventions. Free Radic Biol Med 2025; 227:536-556. [PMID: 39643130 DOI: 10.1016/j.freeradbiomed.2024.12.014] [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: 08/06/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells crucial for initiating and regulating adaptive immune responses, making them promising candidates for therapeutic interventions in various immune-mediated diseases. Increasing evidence suggests that the microenvironment in which cells are cultured, as well as the milieu in which they perform their functions, significantly impact their immunomodulatory properties. Among these environmental factors, the role of oxygen in DC biology and its significance for both their in vitro generation and in vivo therapeutic application require investigation. Unlike the atmospheric oxygen level of 21 % commonly used in in vitro assays, physiological oxygen levels are much lower (3-9 %), and hypoxia (<1.3 %) is a prevalent condition of both healthy tissues and disease states. This mismatch between laboratory and physiological conditions underscores the critical need to culture and evaluate therapeutic cells under physiologically relevant oxygen levels to improve their translational relevance and clinical outcomes. This review explores the characteristic hallmarks of human DCs that are influenced by oxygen-dependent pathways, including metabolism, phenotype, cytokine secretion, and migration. Furthermore, we discuss the potential of manipulating oxygen levels to refine the generation and functionality of DCs for therapeutic purposes.
Collapse
Affiliation(s)
- Antonia Peter
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
44
|
Cohen BE. The Role of the Swollen State in Cell Proliferation. J Membr Biol 2025; 258:1-13. [PMID: 39482485 DOI: 10.1007/s00232-024-00328-x] [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: 08/12/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024]
Abstract
Cell swelling is known to be involved in various stages of the growth of plant cells and microorganisms but in mammalian cells how crucial a swollen state is for determining the fate of the cellular proliferation remains unclear. Recent evidence has increased our understanding of how the loss of the cell surface interactions with the extracellular matrix at early mitosis decreases the membrane tension triggering curvature changes in the plasma membrane and the activation of the sodium/hydrogen (Na +/H +) exchanger (NHE1) that drives osmotic swelling. Such a swollen state is temporary, but it is critical to alter essential membrane biophysical parameters that are required to activate Ca2 + channels and modulate the opening of K + channels involved in setting the membrane potential. A decreased membrane potential across the mitotic cell membrane enhances the clustering of Ras proteins involved in the Ca2 + and cytoskeleton-driven events that lead to cell rounding. Changes in the external mechanical and osmotic forces also have an impact on the lipid composition of the plasma membrane during mitosis.
Collapse
|
45
|
Amissah HA, Antwi MH, Amissah TA, Combs SE, Shevtsov M. More than Just Protein Folding: The Epichaperome, Mastermind of the Cancer Cell. Cells 2025; 14:204. [PMID: 39936995 PMCID: PMC11817126 DOI: 10.3390/cells14030204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
The epichaperome, a dynamic and integrated network of chaperone proteins, extends its roles beyond basic protein folding to protein stabilization and intracellular signal transduction to orchestrating a multitude of cellular processes critical for tumor survival. In this review, we explore the multifaceted roles of the epichaperome, delving into its diverse cellular locations, factors that modulate its formation and function, its liquid-liquid phase separation, and the key signaling and crosstalk pathways it regulates, including cellular metabolism and intracellular signal transduction. We further highlight techniques for isolating and identifying epichaperome networks, pitfalls, and opportunities. Further, we review the profound implications of the epichaperome for cancer treatment and therapy design, underscoring the need for strategic engineering that hinges on a comprehensive insight into the comprehensive structure and workings of the epichaperome across the heterogeneous cell subpopulations in the tumor milieu. By presenting a holistic view of the epichaperome's functions and mechanisms, we aim to underscore its potential as a key target for novel anti-cancer strategies, revealing that the epichaperome is not merely a piece of protein folding machinery but a mastermind that facilitates the malignant phenotype.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Biotechnology, School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia;
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, Winneba CE-122-2486, Central Region, Ghana
| | - Maxwell Hubert Antwi
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana; (M.H.A.); (T.A.A.)
| | - Tawfeek Ahmed Amissah
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana; (M.H.A.); (T.A.A.)
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Saint Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg 197341, Russia
| |
Collapse
|
46
|
Higurashi M, Mori K, Nakagawa H, Uchida M, Ishikawa F, Shibanuma M. Respiratory complex I-mediated NAD + regeneration regulates cancer cell proliferation through the transcriptional and translational control of p21 Cip1 expression by SIRT3 and SIRT7. Mol Oncol 2025. [PMID: 39873399 DOI: 10.1002/1878-0261.13808] [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: 05/17/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 01/30/2025] Open
Abstract
The role of the electron transport chain (ETC) in cell proliferation control beyond its crucial function in supporting ATP generation has recently emerged. In this study, we found that, among the four ETC complexes, the complex I (CI)-mediated NAD+ regeneration is important for cancer cell proliferation. In cancer cells, a decrease in CI activity by RNA interference (RNAi) against NADH:ubiquinone oxidoreductase core subunit V1 (NDUFV1) arrested the cell cycle at the G1/S phase, accompanying upregulation of p21Cip1 cyclin-dependent kinase inhibitor expression. Mechanistically, a decrease in the NAD+/NADH ratio downregulated SIRT3 and SIRT7 function, which suppressed p21Cip1 expression at the translational and transcriptional levels, respectively, resulting in the upregulation of the antiproliferative molecule. Importantly, high expression levels of the core subunits of CI correlated with poor prognosis in patients with the hormone receptor(+)/human epidermal growth factor receptor 2(-) (HR+/HER2-) subtype of breast cancer. Therefore, NDUFV1 and SIRT3/7 have emerged as promising therapeutic targets against this breast cancer subtype.
Collapse
Affiliation(s)
- Masato Higurashi
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Kazunori Mori
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Hidetsugu Nakagawa
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Momoko Uchida
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | | | - Motoko Shibanuma
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, Tokyo, Japan
| |
Collapse
|
47
|
Chi Y, Song C, Jia Q, Zhang R, Sun F, Li Z, Jia Y, An X, Wang Z, Li J. A metal coordination polymer nanoparticle synergistically re-establishes acidosis and enhances chemodynamic therapy for Glioblastoma. Acta Biomater 2025; 192:290-301. [PMID: 39608659 DOI: 10.1016/j.actbio.2024.11.042] [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: 08/17/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Chemodynamic therapy (CDT) has become increasingly important as a tumor treatment strategy, which relies on intracellular acid and hydrogen peroxide to kill tumor cells by generating hydroxyl radicals (·OH) through Fenton/Fenton-like reactions. However, the weakly alkaline intracellular environment considerably caused by the efflux of lactate and H+ from glioblastoma cells is not conducive to CDT performance. Intracellular acidification induced by inhibiting the transmembrane monocarboxylate transporter 4 (MCT4) can enhance the therapeutic efficacy of CDT. Existing approaches suffer from insufficient MCT4 inhibition, involve complex drug synthesis, and have many unsatisfactory side effects. METHODS In this study, we constructed an anti-tumor nanoparticle formed by self-assembly driven by the coordination interaction of Fe3+ and α-cyano-4-hydroxycinnamate (CHC) to avoid safety issues posed by excessive modification. Fe-CHC nanoparticles were designed to decrease intracellular pH through inhibition of MCT4, which transports lactate/H+ to the extracellular space. The resulting intracellular accumulation of lactate and H+ led to fatal acidosis and promoted ·OH generated by Fenton/Fenton-like reactions with the presence of the Fe3+, thus enhancing CDT-induced tumor cell death. RESULTS In vitro and in vivo results revealed that Fe-CHC exerted a significant synergistic anti-tumor effect by re-establishing acidosis and enhancing CDT in glioblastoma. Furthermore, the decreased H+outside the cells caused by the inhibition of lactate/H+ efflux hindered extracellular matrix degradation, thereby inhibiting tumor metastasis. CONCLUSION Fe-CHC is an effective anti-cancer agent against glioblastoma. This study provides valuable insights for developing acid-modulating anti-tumor nanoparticles, as well as enriching and optimizing the application of CDT in tumor therapy. STATEMENT OF SIGNIFICANCE Our study pioneers the Fe-CHC nanoparticle, a metal-coordination polymer that targets MCT4 in glioblastoma cells to restore intracellular acidity and synergize with Fe3+ to boost chemodynamic therapy (CDT). Unlike other studies, Fe3+ and CHC work together to maximize the therapeutic potential and safety of Fe-CHC with minimal complexity. This innovative approach not only increased the production of reactive oxygen species within tumor cells, but also hindered tumor metastasis. Our work has important scientific implications for tumor microenvironment regulation and the application of CDT, and will provide a promising pathway for the treatment of aggressive cancers and attract a wide audience through its scientific implications.
Collapse
Affiliation(s)
- Yajing Chi
- School of Medicine, Nankai University, Tianjin, 300071, China; Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Chaoqi Song
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Qian Jia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China; Guangzhou Institute of Technology, Xidian University, Guangzhou, GuangDong, 510000, China.
| | - Ruili Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Fang Sun
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Zheng Li
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Yuanyuan Jia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China
| | - Xian An
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100071, China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, China.
| | - Jianxiong Li
- School of Medicine, Nankai University, Tianjin, 300071, China; Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
48
|
Hamaguchi R, Elemam NM, Uemoto S, Wada H. Editorial: The impact of alkalizing the acidic tumor microenvironment to improve efficacy of cancer treatment, volume II. Front Oncol 2025; 14:1542787. [PMID: 39876889 PMCID: PMC11772194 DOI: 10.3389/fonc.2024.1542787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Affiliation(s)
- Reo Hamaguchi
- Clinical Cancer Research Team, Japanese Society on Inflammation and Metabolism in Cancer, Kyoto, Japan
| | - Noha Mousaad Elemam
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Hiromi Wada
- Clinical Cancer Research Team, Japanese Society on Inflammation and Metabolism in Cancer, Kyoto, Japan
| |
Collapse
|
49
|
Liu S, Zhang T, Fang L, Hu L, Yin X, Tang X. Integrative pharmacological analysis of modified Zuojin formula: Inhibiting the HIF-1α-mediated glycolytic pathway in chronic atrophic gastritis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119136. [PMID: 39577677 DOI: 10.1016/j.jep.2024.119136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuojin formula (ZJF) is a well-known herbal medicine in Pharmacopoeia of China, which is widely used for gastritis. Modified Zuojin formula (MZJF) was adapted based on traditional Chinese medicine (TCM) theories concerning gastric atrophy and dysplasia, along with extensive clinical experience, has been clinically employed to treat chronic atrophic gastritis (CAG). However, the underlying mechanisms by which MZJF intervenes in CAG remain to be fully elucidated. AIM OF THE STUDY The aim of this study was to evaluate the effects of MZJF intervention in CAG and explore its potential mechanisms. METHODS Four induction factors were used to establish a CAG rat model. HE and AB-PAS staining was utilized to assess the effects of MZJF in the intervention of CAG. The stomach weight index and gastric acid pH was used to assess the overall state of stomach. ELISA was used to assess the gastric mucosal inflammatory response. Using transmission electron microscopy to observe chief cells and parietal cells, we evaluated the improvement of ultrastructure by MZJF. Through network pharmacology analysis, the possible regulatory mechanism of MZJF in CAG was preliminarily explored. Binding interactions between MZJF components and predicted targets were explored using molecular docking. Subsequently, quantitative real-time PCR (qRT-PCR), Western blot, biochemical analysis and TUNEL staining were applied to validate the effect of MZJF on predicted pathway. RESULTS MZJF treatment ameliorated gastric mucosal pathology, inflammation, cellular ultrastructural damage and PG levels, halted the exacerbation of CAG in rats, along with a reduction in stomach weight index and gastric acid pH. A total of 79 compounds in MZJF targeting 203 CAG-related molecules were identified through network pharmacology. Enrichment analysis of the core targets was focused on the hypoxia inducible factor-1α (HIF-1α) signaling pathway. Molecular docking results identified HIF-1α as stable binding targets for MZJF primary components. Subsequently, PCR, WB, and biochemical results showed that MZJF suppressed the gene and protein expression levels of HIF-1α and its downstream molecules including glycolytic enzymes and transporters, modulated glucose, pyruvic acid and lactate levels in gastric mucosal tissue. Moreover, MZJF induced apoptosis of gastric epithelial cells, as evidenced by the upregulation of cleaved caspase-3, Bax, Bax/Bcl-2 and TUNEL positive cells ratio. CONCLUSIONS MZJF suppressed the HIF-1α-mediated glycolytic pathway, and promoted cell apoptosis, thereby halting the malignant transformation of CAG. The study provides a valuable reference point for applying TCM in preventing and treating CAG.
Collapse
Affiliation(s)
- Shan Liu
- Postdoctoral Research Station of China Academy of Chinese Medical Sciences, Institute of Digestive Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Tai Zhang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing, 100091, China; Peking University Health Science Center, Beijing, 100191, China.
| | - Lihui Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Lanshuo Hu
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xiaolan Yin
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361005, China.
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
50
|
Kakehi R, Kobayashi H, Mashiyama H, Yajima T, Koyama H, Ito TK, Yoshida M, Nagaoka Y, Sumiyoshi T. Asymmetric Synthesis, Structure Determination, and Biologic Evaluation of Isomers of TLAM as PFK1 Inhibitors. ACS Med Chem Lett 2025; 16:59-63. [PMID: 39811129 PMCID: PMC11726387 DOI: 10.1021/acsmedchemlett.4c00436] [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: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 01/16/2025] Open
Abstract
Inhibiting phosphofructokinase-1 (PFK1) is a promising approach for treating lactic acidosis and mitochondrial dysfunction by activating oxidative phosphorylation. Tryptolinamide (TLAM) has been shown as a PFK1 inhibitor, but its complex stereochemistry, with 16 possible isomers complicates further development. We conducted an asymmetric synthesis, determined the absolute configurations, and evaluated the PFK1 inhibitory activity of the TLAM isomers. Our structure-activity relationship (SAR) study of TLAM isomers revealed that both carboline and norbornene configurations influence PFK1 inhibitory activity. Among isomers 1a-1d, compound 1c was the most potent PFK1 inhibitor. Our elucidation of the SAR information on PFK1 inhibitors provides valuable insights for effective optimization.
Collapse
Affiliation(s)
- Ryo Kakehi
- Department
of Life Science and Biotechnology, Faculty of Chemistry, Materials
and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita, Osaka 564-8680, Japan
| | - Hiroki Kobayashi
- Laboratory
of Oncology, School of Life Sciences, Tokyo
University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Seed
Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Haruna Mashiyama
- Seed
Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Tatsuo Yajima
- Department
of Chemistry and Materials Engineering, Faculty of Chemistry, Materials
and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita, Osaka 564-8680, Japan
| | - Hiroo Koyama
- Drug
Discovery Platforms Cooperation Division, Drug Discovery Chemistry
Platform Unit, RIKEN Center for Sustainable
Resource Science, 2-1
Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takashi K. Ito
- Chemical
Genomics Research Group, RIKEN Center for
Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Minoru Yoshida
- Seed
Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Chemical
Genomics Research Group, RIKEN Center for
Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Office
of University Professors, The University
of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
- Collaborative
Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yasuo Nagaoka
- Department
of Life Science and Biotechnology, Faculty of Chemistry, Materials
and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita, Osaka 564-8680, Japan
| | - Takaaki Sumiyoshi
- Department
of Life Science and Biotechnology, Faculty of Chemistry, Materials
and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita, Osaka 564-8680, Japan
| |
Collapse
|