1
|
Aghasizadeh M, Moghaddam T, Bahrami AR, Sadeghian H, Alavi SJ, Kazemi T, Matin MM. Evaluation of several farnesyloxycarbostyril derivatives as potential 15-LOX-1 inhibitors for prostate cancer treatment. Toxicol Appl Pharmacol 2025; 498:117293. [PMID: 40057000 DOI: 10.1016/j.taap.2025.117293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
The impact of 15-lipoxygenase-1 (15-LOX-1) in the progression of prostate cancer (PCa) is noteworthy, as it correlates with the Gleason score of the disease. Thus, development of specific 15-LOX-1 inhibitors would be desirable for targeted therapy of PCa. This study focused on evaluating the anti-prostate cancer potency of three farnesyloxycarbostyril derivatives, 6-, 7- and 8-farnesyloxycarbostyril (6-, 7- and 8-FQ), as potential inhibitors of 15-LOX-1 on PCa cells. To this end, the enzymatic activity of 15-LOX was first assessed in PCa and human dermal fibroblast (HDF) cells. Subsequently, the cytotoxic effects and apoptosis-inducing capabilities of the compounds were assessed through MTT assay and FITC-annexin V/PI staining, respectively. Among the compounds, 8-FQ was selected for further assessment in a mouse model bearing xenograft human PCa tumor. The results demonstrated that the most effective compound, 8-FQ, caused an 84-fold and 15.7-fold reduction in 15-LOX activity in PC-3 cells at 30 and 14 μM concentrations, respectively. The MTT assay revealed a dose- and time-dependent toxicity of the compounds on PCa cells, and flow cytometry results indicated that apoptosis served as the dominant mechanism of cell death. Given the upregulation of 15-LOX-1 in human PCa cells, the study concludes that the heightened sensitivity to 8-FQ is likely associated with elevated levels of 15-LOX-1. In vivo experiments using immunosuppressed C57BL/6 mice bearing human PC-3 tumors revealed that 8-FQ, at a dosage of 10 mg/kg, exhibited strong antitumor effects with minimal side effects, indicating its potential as a promising therapeutic agent for PCa following further optimization.
Collapse
Affiliation(s)
- Mehrdad Aghasizadeh
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Tayebe Moghaddam
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Sadeghian
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Jamal Alavi
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tahmineh Kazemi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
2
|
Ghosh D, Guin A, Kumar A, Das A, Paul S. Comprehensive insights of etiological drivers of hepatocellular carcinoma: Fostering targeted nano delivery to anti-cancer regimes. Biochim Biophys Acta Rev Cancer 2025; 1880:189318. [PMID: 40222420 DOI: 10.1016/j.bbcan.2025.189318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as one of the most prevalent and deadliest malignancies on a global scale. Its complex pathogenesis arises from multifactorial etiologies, including viral infections, metabolic syndromes, and environmental carcinogens, all of which drive genetic and molecular aberrations in hepatocytes. This intricate condition is associated with multiple causative factors, resulting in the abnormal activation of various cellular and molecular pathways. Given that HCC frequently manifests within the context of a compromised or cirrhotic liver, coupled with the tendency of late-stage diagnoses, the overall prognosis tends to be unfavorable. Systemic therapy, especially conventional cytotoxic drugs, generally proves ineffective. Despite advancements in therapeutic interventions, conventional treatments such as chemotherapy often exhibit limited efficacy and substantial systemic toxicity. In this context, nanomedicine, particularly lipid-based nanoparticles (LNPs), has emerged as a promising strategy for enhancing drug delivery specificity and reducing adverse effects. This review provides a comprehensive overview of the molecular and metabolic underpinnings of HCC. Furthermore, we explored the role of lipid-based nano-formulations including liposomes, solid lipid nanoparticles, and nanostructured lipid carriers in targeted drug delivery for HCC. We have highlighted recent advances in LNP-based delivery approaches, FDA-approved drugs, and surface modification strategies to improve liver-specific delivery and therapeutic efficacy. It will provide a comprehensive summary of various treatment strategies, recent clinical advances, receptor-targeting strategies and the role of lipid composition in cellular uptake. The review concludes with a critical assessment of existing challenges and future prospects in nanomedicines-driven HCC therapy.
Collapse
Affiliation(s)
- Dipanjan Ghosh
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Aharna Guin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Aryan Kumar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Amlan Das
- Department of Microbiology & Department of Biochemistry, Royal School of Biosciences, The Assam Royal Global University, Guwahati 781035, Assam, India.
| | - Santanu Paul
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India.
| |
Collapse
|
3
|
Karimpur-Zahmatkesh A, Khalaj-Kondori M. The perspective of targeting cancer cell metabolism: combination therapy approaches. Mol Biol Rep 2025; 52:375. [PMID: 40202553 DOI: 10.1007/s11033-025-10472-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells are considered the most adaptable for their metabolic status, which supports growth, survival, rapid proliferation, invasiveness, and metastasis in a nutrient-deficient microenvironment. Since the discovery of altered glucose metabolism (aerobic glycolysis), which is generally known as a part of metabolic reprogramming and an innate trait of cancer cells, in 1930 via Dr. Otto Warburg, numerous studies have endeavored to recognize various aspects of cancer cell metabolism and find new methods for efficiently eradicating described cells by targeting their energy metabolism. In this way, the outcomes have mainly been promising. Accordingly, outlining the related results will indeed assist us in making a definitive path for developing targeted therapy strategies based on cancer cell-altered metabolism. The present study reviews the key features of cancer cell metabolism and treatment strategies based on them. It emphasizes the importance of targeting cancer cell dysregulated metabolic pathways that influence the cell energy supply and manage cancer cell growth and survival. This trial also introduces a multimodal therapeutic strategy hypothesis, a potential next-generation combination therapy approach, and suggests interdisciplinary research to recognize the complexities of cancer metabolism and exploit them for designing more efficacious cancer therapeutic strategies.
Collapse
Affiliation(s)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
4
|
Xiao X, Liu Y, Qu X, Liu L, Li GQ, Chen H, Zhou L, Liu Y. Aryl hydrocarbon receptor-regulated long non-coding RNAs: implications for glycolipid metabolism and prognosis in hepatocellular carcinoma. Front Oncol 2025; 15:1537481. [PMID: 40248203 PMCID: PMC12003141 DOI: 10.3389/fonc.2025.1537481] [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: 11/30/2024] [Accepted: 02/21/2025] [Indexed: 04/19/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths with limited treatment options. Tumor metabolic disorder is elevated in HCC and activates the aryl hydrocarbon receptor (AHR), a transcription factor implicated in cancer progression. However, the role of AHR in regulating long non-coding RNAs (lncRNAs) and their impact on glycolipid metabolism remains underexplored. Materials and methods We investigated AHR's influence on several HCC cell lines treated with the AHR ligand. RNA sequencing was performed to identify the differentially expressed (DE) lncRNAs and mRNAs. We analyzed the differences and then conducted functional pathway enrichment of the identified DE lncRNAs and mRNAs. Furthermore, we constructed co-expression networks of lncRNAs and mRNAs and performed survival analysis using The Cancer Genome Atlas (TCGA) data. Results RNA sequencing identified a substantial number of lncRNAs and mRNAs. DEG analysis identified the significant differences between them related to cancer progression, with pathways such as PI3K-Akt, VEGF, and PPAR signaling highlighted. A co-expression network was utilized to elucidate the lncRNA-mRNA interactions and their regulation of glycolipid metabolism.Survival analysis identified the AHR-regulated lncRNAs associated with poor prognosis, like ASAP1-IT1 and RMDN2-AS1. Conclusion This study clarifies AHR's role in regulating gene expression and metabolism in HCC, revealing novel lncRNA biomarkers and potential therapeutic targets that could aid HCC. Further research is needed to explore AHR's effects on the regulation of glucose-lipid metabolism in HCC.
Collapse
Affiliation(s)
- Xiaoli Xiao
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yao Liu
- Department of Gastroenterology, the First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China
| | - Xiaoyong Qu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo-Qing Li
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Honghui Chen
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linlin Zhou
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yanping Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
5
|
Sobolev V, Tchepourina E, Soboleva A, Denisova E, Korsunskaya I, Mezentsev A. PPAR-γ in Melanoma and Immune Cells: Insights into Disease Pathogenesis and Therapeutic Implications. Cells 2025; 14:534. [PMID: 40214488 PMCID: PMC11989151 DOI: 10.3390/cells14070534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Changes in skin pigmentation, like hyperpigmentation or moles, can affect appearance and social life. Unlike locally containable moles, malignant melanomas are aggressive and can spread rapidly, disproportionately affecting younger individuals with a high potential for metastasis. Research has shown that the peroxisome proliferator-activated receptor gamma (PPAR-γ) and its ligands exhibit protective effects against melanoma. As a transcription factor, PPAR-γ is crucial in functions like fatty acid storage and glucose metabolism. Activation of PPAR-γ promotes lipid uptake and enhances sensitivity to insulin. In many cases, it also inhibits the growth of cancer cell lines, like breast, gastric, lung, and prostate cancer. In melanoma, PPAR-γ regulates cell proliferation, differentiation, apoptosis, and survival. During tumorigenesis, it controls metabolic changes and the immunogenicity of stromal cells. PPAR-γ agonists can target hypoxia-induced angiogenesis in tumor therapy, but their effects on tumors can be suppressive or promotional, depending on the tumor environment. Published data show that PPAR-γ-targeting agents can be effective in specific groups of patients, but further studies are needed to understand lesser-known biological effects of PPAR-γ and address the existing safety concerns. This review provides a summary of the current understanding of PPAR-γ and its involvement in melanoma.
Collapse
Affiliation(s)
- Vladimir Sobolev
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Ekaterina Tchepourina
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Anna Soboleva
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Elena Denisova
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
- Moscow Center of Dermatovenerology and Cosmetology, Moscow 119071, Russia
| | - Irina Korsunskaya
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Alexandre Mezentsev
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| |
Collapse
|
6
|
Zhang C, Li M, Yang M, Lin J, Huang J, Lin Y, Chen X, Liang Y, Yang Y, Yu Z, Hu D, Zhang M, Hu F. Plasma metabolites, systolic blood pressure, lifestyle, and stroke risk: A prospective cohort study. Int J Stroke 2025; 20:486-496. [PMID: 39394735 DOI: 10.1177/17474930241293408] [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] [Indexed: 10/14/2024]
Abstract
BACKGROUND To estimate the associations of stroke risk with plasma metabolites, metabolic risk score (MRS), the combinations of MRS with hypertension or lifestyle, and lifestyle-related metabolic signature. To assess the improvement of the stroke risk prediction model through the incorporation of MRS. METHODS A total of 77,315 participants from the UK Biobank were included in this study. Xgboost and LASSO-Cox regression were used to select metabolites and construct MRS. Elastic net regression was utilized to construct the lifestyle-related metabolic signature. Multivariate Cox regression was used to estimate the associations between metabolites, MRS, the combinations of MRS with hypertension or lifestyle, lifestyle-related metabolic signature, and stroke risk. RESULTS We identified 48, 63, 39, and 4 metabolites associated with the risk of stroke, ischemic stroke (IS), subarachnoid hemorrhage (SAH), and intracerebral hemorrhage (ICH), respectively. High MRS significantly increased the risk of stroke (HR = 2.65 (95% confidence interval (CI): 2.09-3.35)), IS (HR = 2.45 (95% CI: 1.89-3.17)), ICH (HR = 2.74 (95% CI: 1.55-4.85)), and SAH (HR = 4.64 (95% CI: 2.25-9.56)). In the combination analyses, compared with normal systolic blood pressure (SBP) and low MRS, normal/high SBP, and high MRS significantly increased stroke risk (HR = 5.80 (95% CI: 2.75-12.27)/6.37 (95% CI: 3.22-12.62)). A favorable/unfavorable lifestyle and high MRS also significantly increased stroke risk (HR = 2.38 (95% CI: 1.73-3.28)/3.86 (95% CI: 2.63-5.67)) compared with a favorable lifestyle and low MRS. Incorporating MRS into the 15-year stroke and IS risk prediction model increased the areas under the curves (AUCs) from 0.746 to 0.766 and from 0.771 to 0.811, respectively. The metabolic signature was correlated with adherence to a healthy lifestyle (r = 0.414; P = 2.22e-16) and inversely associated with stroke risk (HR = 0.80 (95% CI: 0.73-0.86)). CONCLUSIONS Various metabolites and MRS were significantly associated with the risk of stroke, IS, ICH, and SAH. Individuals with a high MRS may face an elevated stroke risk among populations with high SBP or unhealthy lifestyle, even those with normal SBP or healthy lifestyle. MRS provided modest improvement to the stroke risk prediction model. The lifestyle-related metabolic signature could reduce 20% stroke risk.
Collapse
Affiliation(s)
- Canjia Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- 2019 Preventive Medicine, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Mingxiao Li
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- 2022 Preventive Medicine, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Miaomiao Yang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Jiaqi Lin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Jinyao Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Ying Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Xi Chen
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yongqiang Liang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- 2019 Preventive Medicine, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yuanhai Yang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- 2020 Preventive Medicine, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Ziyuan Yu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- 2020 Preventive Medicine, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| |
Collapse
|
7
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
8
|
Wang L, Wei L, Chen X, Xiong J. Arachidonic acid suppresses lung cancer cell growth and modulates lipid metabolism and the ERK/PPARγ signaling pathway. Lipids Health Dis 2025; 24:114. [PMID: 40140862 PMCID: PMC11948643 DOI: 10.1186/s12944-025-02490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality worldwide, necessitating the development of new treatment strategies. Arachidonic acid (ARA), a polyunsaturated fatty acid, shows promise in cancer therapy due to its potential anti-tumor effects, although its role in lung cancer remains unclear. This study investigated the effects and underlying mechanism of ARA on A549 and NCI-H1299 lung cancer cells. In vitro assays were used to assess cell viability, apoptosis, colony formation, lipid droplet formation, and changes in cellular lipid content. ARA dose-dependently suppressed cell viability, facilitated apoptosis, and suppressed colony formation in both lung cancer cell lines. Network pharmacology analysis was performed to identify potential gene targets and pathways, uncovering 61 overlapping genes between ARA and lung cancer-related targets, with mitogen-activated protein kinase 1 (MAPK1) emerging as a key gene. Enrichment analyses suggested that the effects of ARA might be mediated through lipid metabolism and the extracellular signal-regulated kinase (ERK)/peroxisome proliferator-activated receptor gamma (PPARγ) signaling pathway. In both lung cancer cell lines, ARA treatment inhibited lipid droplet formation and decreased the cellular lipids. Immunoblotting further confirmed that ARA treatment significantly increased ERK phosphorylation while reducing PPARγ and fatty acid synthase (FASN) protein levels. In vitro experiments using GW9662, a PPARγ antagonist, confirmed that inhibiting lipid droplet formation impairs lung cancer cell viability and promotes apoptosis. Furthermore, in vivo experiments demonstrated that ARA significantly reduced tumor size and weight in a lung cancer xenograft model, further validating its anti-tumor effects. The potential of ARA as a therapeutic agent for lung cancer might involve lipid metabolism and relevant signaling pathways. A future study exploring the full therapeutic potential of ARA and underlying mechanisms in lung cancer is needed.
Collapse
Affiliation(s)
- Lin Wang
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, ZhuZhou, 412000, Hunan Province, China
| | - Lanlan Wei
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, ZhuZhou, 412000, Hunan Province, China
| | - Xueling Chen
- Emergency Center, Zhuzhou Central Hospital, ZhuZhou, 412000, Hunan Province, China
| | - Jiali Xiong
- Department of Respiratory and Critical Care Medicine, Zhuzhou Central Hospital, ZhuZhou, 412000, Hunan Province, China.
| |
Collapse
|
9
|
Heravi G, Liu Z, Herroon M, Wilson A, Fan YY, Jiang Y, Vakeesan N, Tao L, Peng Z, Zhang K, Li J, Chapkin RS, Podgorski I, Liu W. Targeting polyunsaturated fatty acids desaturase FADS1 inhibits renal cancer growth via ATF3-mediated ER stress response. Biomed Pharmacother 2025; 186:118006. [PMID: 40121894 DOI: 10.1016/j.biopha.2025.118006] [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: 12/13/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVE Fatty Acid Desaturase 1 (FADS1) is a rate-limiting enzyme controlling the bioproduction of long-chain polyunsaturated fatty acids (PUFAs). Increasing studies suggest that FADS1 is a potential cancer target. Our previous research has demonstrated the significant role of FADS1 in cancer biology and patient survival, especially in kidney cancers. We aim to explore the underlying mechanism in this study. METHOD AND RESULTS We found that pharmacological inhibition or knockdown of the expression of FADS1 significantly reduced the intracellular conversion of long-chain PUFAs, effectively inhibits renal cancer cell proliferation, and induces cell cycle arrest. The stable knockdown of FADS1 also significantly inhibits tumor formation in vivo. Mechanistically, we showed that while FADS1 inhibition induces endoplasmic reticulum (ER) stress, FADS1 expression is augmented by ER-stress inducer, suggesting a necessary role of PUFA production in response to ER stress. FADS1-inhibition sensitized cellular response to ER stress inducers, leading to cell apoptosis. Also, FADS1 inhibition-induced ER stress leads to activation of the PERK/eIF2α/ATF4/ATF3 pathway. Inhibiting PERK or knockdown of ATF3 rescued FADS1 inhibition-induced ER stress and cell growth suppression, while ATF3-overexpression aggravates the FADS1 inhibition-induced cell growth suppression and leads to cell death. Metabolomic analysis revealed that FADS1 inhibition results in decreased level of UPD-N-Acetylglucosamine, a critical mediator of the unfolded protein response, as well as impaired biosynthesis of nucleotides, possibly accounting for the cell cycle arrest. CONCLUSION Our findings suggest that PUFA desaturation is crucial for rescuing cancer cells from persistent ER stress, supporting FADS1 as a new therapeutic target.
Collapse
Affiliation(s)
- Gioia Heravi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Zhenjie Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Mackenzie Herroon
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Alexis Wilson
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Yang-Yi Fan
- Department of Nutrition, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843, USA
| | - Yang Jiang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Nivisa Vakeesan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Li Tao
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Jing Li
- Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Robert S Chapkin
- Department of Nutrition, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843, USA; CPRIT Regional Center of Excellence in Cancer Research, Texas A&M University, College Station, TX 77843, USA
| | - Izabela Podgorski
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; Department of Oncology, School of Medicine, Wayne State University, and Karmanos Cancer Institute, Detroit, MI 48201, USA.
| |
Collapse
|
10
|
Hossain MK, Unger L, Larsen U, Altankhuyag A, Legøy TA, Paulo JA, Vethe H, Ghila L. Mapping the initial effects of carcinogen-induced oncogenic transformation in the mouse bladder. Exp Cell Res 2025; 446:114452. [PMID: 39988124 DOI: 10.1016/j.yexcr.2025.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Characterizing the initial stages of oncogenic transformation allows the identification of tumor-promoting processes before the inherent clonal selection of the aggressive clones. Here, we used global proteomics, genetic cell tracing, and immunofluorescence to dynamically map the very early stages of cancer initiation in a mouse model of bladder cancer. We observed a very rapid and incremental proteome dysregulation, with changes in the energy metabolism, proliferation and immune signatures dominating the landscape. The changes in the lipid metabolism were immediate and defined by an increase fatty acid metabolism and lipid transport, followed by the activation of the immune landscape. Alongside the changes in the immune signature and lipid metabolism, we also mapped a clear increase in the cell cycle-related pathways and proliferation. Proliferation was mainly restricted to the basal epithelial layer rapidly leading to urothelium thickening, despite the progressive loss of the superficial layer. Moreover, we observed a tilt in the energy balance towards increased glucose metabolism, probably characterizing cells of the tumor microenvironment. All of the observed proteome signature changes were persistent, being retained and sometimes intensified or diversified along the timeline. The signatures observed in this pilot suggest these processes as potentially targetable drivers of the future neoplastic transformations in the bladder.
Collapse
Affiliation(s)
- Md Kaykobad Hossain
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Lucas Unger
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Ulrik Larsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | | | - Thomas Aga Legøy
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heidrun Vethe
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Norway
| | - Luiza Ghila
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway.
| |
Collapse
|
11
|
Phulara NR, Ishida CT, Espenshade PJ, Seneviratne HK. Gemcitabine Alters Phosphatidylcholine Metabolism in Mouse Pancreatic Tumors. J Proteome Res 2025; 24:1209-1218. [PMID: 39973059 DOI: 10.1021/acs.jproteome.4c00839] [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] [Indexed: 02/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest diseases, despite advancements in elucidating tumor biology and developing novel therapeutics. Importantly, lipids, such as phospholipids, are crucial for the survival and proliferation of tumor cells. However, the impact of chemotherapeutic drugs on phospholipid metabolism in PDAC remains poorly understood. Gemcitabine (a nucleoside analogue) is a first-line drug in PDAC treatment, but its clinical effectiveness is limited by multiple factors. Herein, we employed matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) and proteomics approaches to investigate gemcitabine-induced lipid metabolism alterations in mouse pancreatic tumors following gemcitabine treatment (n = 3, control tumors; n = 3, gemcitabine-treated tumors). From MALDI MSI experiments, we observed elevated levels of several phosphatidylcholines (PCs), PC(30:0), PC(32:3), PC(34:2), PC(36:1), and PC(36:2), in gemcitabine-treated tumor tissues compared to the control. In addition, proteomics data revealed the differential abundance of several phospholipid-binding proteins in response to gemcitabine treatments. Furthermore, several endoplasmic reticulum stress-related proteins exhibited high expression in gemcitabine-treated tumor tissues. Altogether, our MALDI MSI and proteomics data provide important insights into alterations in PC metabolism in pancreatic tumors in response to gemcitabine treatment. Importantly, targeting the altered PC metabolism during gemcitabine therapy might help combat pancreatic cancer.
Collapse
Affiliation(s)
- Nav Raj Phulara
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Chiaki Tsuge Ishida
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Peter John Espenshade
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Giovanis Institute for Translational Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
12
|
Cai X, Li X, Zhang M, Dong Z, Weng Y, Yu W. RBM15 promotes lipogenesis and malignancy in gastric cancer by regulating N6-Methyladenosine modification of ACLY mRNA in an IGF2BP2-dependent manner. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159580. [PMID: 39549859 DOI: 10.1016/j.bbalip.2024.159580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
N6-methyladenosine (m6A) and lipid metabolism reprogramming play pivotal roles in cancer development. Nevertheless, the precise functions of m6A methyltransferase RNA Binding Motif Protein 15 (RBM15) and its interactions with ATP Citrate Lyase (ACLY) in gastric cancer (GC) have not been fully elucidated. In this study, we comprehensively investigate the biological roles and potential mechanisms of RBM15 and ACLY in GC. We employed a combination of fundamental experiments and bioinformatics analyses to unravel the enigmatic roles of RBM15 and ACLY. The expression of RBM15 was evaluated. The biological roles of RBM15 in GC cells were investigated through in vitro and in vivo studies. ACLY was selected as the candidate target of RBM15. Subsequently, to decipher the underlying mechanisms of the RBM15/ACLY axis, we conducted a series of experiments including methylated RNA immunoprecipitation qPCR, dual-luciferase reporter assays, and RNA immunoprecipitation qPCR. We observed a conspicuous upregulation of RBM15 in GC, and its heightened expression was associated with an unfavorable prognosis. Functionally, RBM15 fostered the proliferation and invasiveness of GC cells both in vitro and in vivo. Mechanistically, ACLY emerged as the downstream target of RBM15 and it was validated as an oncogene in GC cells. RBM15 mediated the activation of ACLY by regulating m6A modification in an IGF2BP2-dependent manner, thereby driving lipogenesis and exacerbating the malignant characteristics in GC. The activation of ACLY, facilitated by RBM15/IGF2BP2-mediated m6A modification, drives lipogenesis and promotes the progression of GC.
Collapse
Affiliation(s)
- Xianlei Cai
- Department of Gastrointestinal Surgery, The Lihuili Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China.
| | - Xueying Li
- Department of Gastroenterology, The First Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China
| | - Miaozun Zhang
- Department of Gastrointestinal Surgery, The Lihuili Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China
| | - Zhebin Dong
- Department of Gastrointestinal Surgery, The Lihuili Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China
| | - Yihui Weng
- Department of Gastrointestinal Surgery, The Lihuili Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China
| | - Weiming Yu
- Department of Gastrointestinal Surgery, The Lihuili Affiliated Hospital, Ningbo University, Ningbo, Zhejiang 315000, China.
| |
Collapse
|
13
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [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: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
14
|
Choe S, Jeon M, Yoon H. Advanced Therapeutic Approaches for Metastatic Ovarian Cancer. Cancers (Basel) 2025; 17:788. [PMID: 40075635 PMCID: PMC11898553 DOI: 10.3390/cancers17050788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death among women, which is one of the most common gynecological cancers worldwide. Although several cytoreductive surgeries and chemotherapies have been attempted to address ovarian cancer, the disease still shows poor prognosis and survival rates due to prevalent metastasis. Peritoneal metastasis is recognized as the primary route of metastatic progression in ovarian cancer. It causes severe symptoms in patients, but it is generally difficult to detect at an early stage. Current anti-cancer therapy is insufficient to completely treat metastatic ovarian cancer due to its high rates of recurrence and resistance. Therefore, developing strategies for treating metastatic ovarian cancer requires a deeper understanding of the tumor microenvironment (TME) and the identification of effective therapeutic targets through precision oncology. Given that various signaling pathways, including TGF-β, NF-κB, and PI3K/AKT/mTOR pathways, influence cancer progression, their activity and significance can vary depending on the cancer type. In ovarian cancer, these pathways are particularly important, as they not only drive tumor progression but also impact the TME, which contributes to the metastatic potential. The TME plays a critical role in driving metastatic features in ovarian cancer through altered immunologic interactions. Recent therapeutic advances have focused on targeting these distinct features to improve treatment outcomes. Deciphering the complex interaction between signaling pathways and immune populations contributing to metastatic ovarian cancer provides an opportunity to enhance anti-cancer efficacy. Hereby, this review highlights the mechanisms of signaling pathways in metastatic ovarian cancer and immunological interactions to understand improved immunotherapy against ovarian cancer.
Collapse
Affiliation(s)
- Soohyun Choe
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Minyeong Jeon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| |
Collapse
|
15
|
Jin Z, Chen Y. Serum PM20D1 levels are associated with nutritional status and inflammatory factors in gastric cancer patients undergoing early enteral nutrition. Open Med (Wars) 2025; 20:20241111. [PMID: 39927167 PMCID: PMC11806234 DOI: 10.1515/med-2024-1111] [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: 05/21/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 02/11/2025] Open
Abstract
Background and objective Early nutritional support holds paramount importance for postoperative gastric cancer (GC) patients. Peptidase M20 domain containing 1 (PM20D1) is a secretory enzyme associated with glucose and lipid metabolism. However, there is a dearth of clinical studies delving into the connection between PM20D1, lipid metabolism, and inflammatory factors in GC patients who have received enteral nutrition (EN). This research aimed to investigate the serum levels of PM20D1 in GC patients following early EN and their potential associations with lipid metabolism, nutritional markers, and inflammatory factors. Methods This prospective observational study enrolled 180 GC patients between May 2020 and July 2022. On the first postoperative day, all patients received EN support, which was maintained for a duration of 5 days. Serum levels of PM20D1, interleukin (IL)-6, IL-1β, and C-reactive protein were measured on the sixth day after surgery using an enzyme-linked immunosorbent assay. Data on demographics, clinical statistics, lipid metabolism, nutritional parameters, and the prognostic nutritional index (PNI) were collected. Patients were followed up for 12 months, and both overall survival and disease-free survival were recorded. Results In the low PNI group, the serum levels of PM20D1, albumin (ALB), and blood lymphocytes (BL) showed significant reductions. Pearson analysis revealed a negative correlation between PM20D1 and IL-6 levels, whereas a positive correlation emerged between PM20D1 and ALB and BL levels. Furthermore, PM20D1 demonstrated potential as a biomarker for diagnosing poor nutritional status (PNI < 43) in GC patients and was a risk factor for poor nutritional status in GC patients. Conclusion Serum PM20D1 remarkably declined in GC patients after early EN and was associated with poor nutritional status.
Collapse
Affiliation(s)
- Zhengyu Jin
- Department of General Surgery, Suzhou Dushu Lake Hospital,
Suzhou, 215000, Jiangsu, P. R. China
| | - Yuning Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Street, Tianning District, Changzhou, 213000, Jiangsu, P. R. China
| |
Collapse
|
16
|
Liu S, Liu Z, Lei H, Miao YB, Chen J. Programmable Nanomodulators for Precision Therapy, Engineering Tumor Metabolism to Enhance Therapeutic Efficacy. Adv Healthc Mater 2025; 14:e2403019. [PMID: 39529548 DOI: 10.1002/adhm.202403019] [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: 08/13/2024] [Revised: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Tumor metabolism is crucial in the continuous advancement and complex growth of cancer. The emerging field of nanotechnology has made significant strides in enhancing the understanding of the complex metabolic intricacies inherent to tumors, offering potential avenues for their strategic manipulation to achieve therapeutic goals. This comprehensive review delves into the interplay between tumor metabolism and various facets of cancer, encompassing its origins, progression, and the formidable challenges posed by metastasis. Simultaneously, it underscores the classification of programmable nanomodulators and their transformative impact on enhancing cancer treatment, particularly when integrated with modalities such as chemotherapy, radiotherapy, and immunotherapy. This review also encapsulates the mechanisms by which nanomodulators modulate tumor metabolism, including the delivery of metabolic inhibitors, regulation of oxidative stress, pH value modulation, nanoenzyme catalysis, nutrient deprivation, and RNA interference technology, among others. Additionally, the review delves into the prospects and challenges of nanomodulators in clinical applications. Finally, the innovative concept of using nanomodulators to reprogram metabolic pathways is introduced, aiming to transform cancer cells back into normal cells. This review underscores the profound impact that tailored nanomodulators can have on tumor metabolic, charting a path toward pioneering precision therapies for cancer.
Collapse
Affiliation(s)
- Siwei Liu
- Women & Children's Molecular Medicine Center, Department of Gynecology, Guangyuan Central Hospital, No. 16, Jingxiangzi, Lizhou District, Guangyuan, 628000, P. R. China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Huajiang Lei
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Jiao Chen
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| |
Collapse
|
17
|
Jog E, Jainarayanan AK, La Ferlita A, Chakraborty A, Dalwai A, Yahya S, Shivashankar A, Choudhary BS, Chandramouli A, Kazi M, Jain D, Khapare N, B A, Khan BK, Gera P, Patil P, Thorat R, Verma N, Sehgal L, Saklani A, Kamat SS, Dalal SN, Chaudhary N. Inhibiting de novo lipogenesis identifies a therapeutic vulnerability in therapy-resistant colorectal cancer. Redox Biol 2025; 79:103458. [PMID: 39705849 PMCID: PMC11729006 DOI: 10.1016/j.redox.2024.103458] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
A significant clinical challenge in patients with colorectal cancer (CRC), which adversely impacts patient survival, is the development of therapy resistance leading to a relapse. Therapy resistance and relapse in CRC is associated with the formation of lipid droplets (LD) by stimulating de novo lipogenesis (DNL). However, the molecular mechanisms underlying the increase in DNL and the susceptibility to DNL-targeted therapies remain unclear. Our study demonstrates that colorectal drug-tolerant persister cells (DTPs) over-express Lipin1 (LPIN1), which facilitates the sequestration of free fatty acids into LDs. The increased expression is mediated by the ETS1-PTPN1-c-Src-CEBPβ pathway. Blocking the conversion of free fatty acids into LDs by treatment with statins or inhibiting lipin1 expression disrupts lipid homeostasis, leading to lipotoxicity and ferroptotic cell death in both DTPs and patient-derived organoids (PDOs) in vitro. Ferroptosis inhibitors or N-acetylcysteine (NAC) can alleviate lipid ROS and cell death resulting from lipin1 inhibition. This strategy also significantly reduces tumor growth in CRC DTP mouse xenograft and patient-derived xenograft (PDX) models. Our findings highlight a new metabolic vulnerability in CRC DTPs, PDO, and PDX models and provide a framework for the rational repurposing of statins. Targeting the phosphatidic acid (PA) to diacylglycerol (DAG) conversion to prevent lipid droplet formation could be an effective therapeutic approach for therapy-resistant CRC.
Collapse
Affiliation(s)
- Eeshrita Jog
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Ashwin Kumar Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
| | - Alessandro La Ferlita
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Arnab Chakraborty
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Afiya Dalwai
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Showket Yahya
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Anusha Shivashankar
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Bhagya Shree Choudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Aakash Chandramouli
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Mufaddal Kazi
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Darshan Jain
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Nileema Khapare
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Akshaya B
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Bushra K Khan
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Poonam Gera
- Biorepository, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Prachi Patil
- Department of Digestive Disease and Clinical Nutrition India, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Nandini Verma
- TNBC Precision Medicine Group, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Lalit Sehgal
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA; Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Avanish Saklani
- Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Department of Gastrointestinal Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai 400012, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune, Maharashtra, 411008, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Nazia Chaudhary
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.
| |
Collapse
|
18
|
Liu Q, Yang R, Wang D, Liu Q. Role of low-density cholesterol and Interleukin-17 interaction in breast cancer pathogenesis and treatment. Cell Biol Int 2025; 49:139-153. [PMID: 39318044 DOI: 10.1002/cbin.12250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Breast cancer (BC) has become the most prevalent cancer worldwide, and further research is being conducted to deepen our understanding of its pathogenesis and treatment. Lipid metabolism disorder is a significant alteration in cancer cells, and the investigation into the role of Interleukin-17 (IL-17) in malignant tumors has emerged as a research focus in recent years. Thus, exploring changes in lipid metabolism and inflammatory factors in BC cells is crucial in identifying potential therapeutic targets. This article summarizes the progress made in the research on the main low-density cholesterol (LDL) transporter and IL-17 in lipid metabolism, and their potential involvement in the development of BC. The article aims to establish a theoretical foundation for the development of BC-related therapies.
Collapse
Affiliation(s)
- Qingqing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Rongyuan Yang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Dawei Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
- The 1st Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangdong, 510405, China
| | - Qing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| |
Collapse
|
19
|
Lin YH, Chen CY, Chi HC, Wu MH, Lai MW, Yeh CT. ANGPTL3 overcomes sorafenib resistance via suppression of SNAI1 and CPT1A in liver cancer. Transl Oncol 2025; 52:102250. [PMID: 39708716 PMCID: PMC11732165 DOI: 10.1016/j.tranon.2024.102250] [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: 04/18/2024] [Revised: 11/14/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024] Open
Abstract
Liver cancer, encompassing hepatocellular carcinoma (HCC) and hepatoblastoma, the latter of which primarily occurs in early childhood, is the most common malignant tumor arising from liver and is responsible for a significant number of cancer-related deaths worldwide. Targeted drugs have been used for anti-liver cancer treatment in the advanced stage, while their efficacy is greatly compromised by development of drug resistance. Drug resistance is a complicated process regulated by intrinsic and extrinsic signals and has been associated with poorer prognosis in cancer patients. In the current study, online available dataset analysis uncovered that angiopoietin-like protein 3 (ANGPTL3) manifested lower expression in sorafenib-resistant liver cancer cell lines. Additionally, ANGPTL3 was downregulated in HCC tissues, with its expression positively correlated with good prognosis. Functionally, ectopic expression of ANGPTL3 re-sensitized sorafenib-resistant cells, enhancing the sorafenib-induced reduction in cell viability and migration by suppressing zinc finger protein SNAI1 (SNAI1) expression and the protein stability of carnitine O-palmitoyltransferase 1, liver isoform (CPT1A). Clinical correlation analysis revealed that ANGPTL3 was negatively associated with SNAI1 expression. In conclusion, we identify a novel association between ANGPTL3, SNAI1 and CPT1A on sorafenib therapeutic response. Targeting ANGPTL3/SNAI1/CPT1A axis may serve as a therapeutic approach to improve prognosis of liver cancer patients with sorafenib resistance.
Collapse
Affiliation(s)
- Yang-Hsiang Lin
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Cheng-Yi Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Hsiang-Cheng Chi
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.
| | - Meng-Han Wu
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Ming-Wei Lai
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; Department of Pediatrics, Chang Gung Memorial Hospital, Linkou Branch and Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.
| |
Collapse
|
20
|
HARISA GAMALELDINI, ALZHRANI RIYADF, ALLUHAIDAN ABDULRAHMANA, ALAMRI SULTANM, BAKHEIT AHMEDH, ASIRI HANADIH, ATTIA SABRYM. Chitosan capped-NLCs enhanced codelivery of gefitinib and simvastatin into MDR HCC: impact of compositions on cell death, JNK3, and Telomerase. Oncol Res 2025; 33:477-492. [PMID: 39866231 PMCID: PMC11754001 DOI: 10.32604/or.2024.053337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/08/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a health problem due to multi-drug resistance (MDR). Codelivery of multiple oncotherapy in one cargo as chimeric cancer therapy (CCT) is suggested as a solution for MDR. This study aims to engineer chitosan-coated nanostructure lipid carriers (NLCs) loaded with gefitinib (GF) and simvastatin (SV) as CCT for HCC. Methods Both GF and SV-loaded nanostructure lipids carriers (GFSVNLC) and chitosan-capped GF and SV-loaded nanostructure lipids carriers (CGFSVNLC) formulations were assembled by top-down techniques. Moreover, particle size (PS), zeta potential (ZP), and polydispersity index (PDI) were measured by Zetasizer. The biosafety of GFSVNLC preparations was investigated by using erythrocytes as a biological model. The cytotoxic, and apoptotic effects of the prepared GFSVNLCs were investigated using HepG2 cell lines as a substitute model for HCC. The effect of GF, SV, and NLC composition on JNK3, HDAC6, and telomerase was studied using molecular docking simulation (MDS). Results The present results revealed that the obtained GFSVNLC and CGFSVNLC have nanosized and consistent, CS coating shifts anionic ZP of GFSVNLC into CGFSVNLC with cationic ZP. Moreover, both formulations are biocompatible as indicated by their gentle effect on erythrocyte hemolysis. The treatment of HepG2 cells with GFSVNLC, and CGFSVNLC induced marked cell death compared to other groups with a decrease of IC50. Equally, the percentage of the apoptotic HepG2 cells was increased upon treatment of the cells with GFSV, GFSVNLC, and CGFSVNLC compared to the control group. Additionally, GF, SV, stearic acid (SA), and oleic acid (OA) modulate the activity of JNK3, HDAC6, and telomerase. Conclusions This study suggests CGFSVNLC achieves codelivery, selective targeting, and enhancing the synergistic effect of GF and SV for inducing HepG2 cell death. Mechanistically, CGFSVNLC inhibits key cascades implicated in MDR and HepG2 cell survival. CGFSVNLC is promising for overcoming drug resistance mechanisms and improving therapeutic outcomes against HepG2 cells.
Collapse
Affiliation(s)
- GAMALELDIN I. HARISA
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - RIYAD F. ALZHRANI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | | | - SULTAN M. ALAMRI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - AHMED H. BAKHEIT
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - HANADI H. ASIRI
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - SABRY M. ATTIA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
21
|
Qian W, Tang H, Yao H. Lipidomics and temporal-spatial distribution of organelle lipid. J Biol Methods 2025; 12:e99010049. [PMID: 40200947 PMCID: PMC11973048 DOI: 10.14440/jbm.2025.0094] [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: 10/08/2024] [Revised: 11/17/2024] [Accepted: 12/16/2024] [Indexed: 04/10/2025] Open
Abstract
Background Lipids are crucial signaling molecules or cellular membrane components orchestrating biological processes. To gain insights into lipid functions and the communication between organelles, it is essential to understand the subcellular localization of individual lipids. Advancements in lipid quantification techniques, improvements in chemical and spatial resolution for detecting various lipid species, and enhancements in organelle isolation speed have allowed for profiling of the organelle lipidome, capturing its temporal-spatial distribution. Objective This review examined approaches used to develop organelle lipidome and aimed to gain insights into cellular lipid homeostasis from an organelle perspective. In addition, this review discussed the advancements in lipid-mediated inter-organelle communication within complex physiological and pathological processes. Conclusion With the advancement of lipidomic technologies, more detailed explorations of organelle structures and the specific lipid-mediating functions they perform are feasible.
Collapse
Affiliation(s)
- Wenjuan Qian
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hongyan Yao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
22
|
Mendoza EN, Ciriolo MR, Ciccarone F. Hypoxia-Induced Reactive Oxygen Species: Their Role in Cancer Resistance and Emerging Therapies to Overcome It. Antioxidants (Basel) 2025; 14:94. [PMID: 39857427 PMCID: PMC11762716 DOI: 10.3390/antiox14010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Normal tissues typically maintain partial oxygen pressure within a range of 3-10% oxygen, ensuring homeostasis through a well-regulated oxygen supply and responsive vascular network. However, in solid tumors, rapid growth often outpaces angiogenesis, creating a hypoxic microenvironment that fosters tumor progression, altered metabolism and resistance to therapy. Hypoxic tumor regions experience uneven oxygen distribution with severe hypoxia in the core due to poor vascularization and high metabolic oxygen consumption. Cancer cells adapt to these conditions through metabolic shifts, predominantly relying on glycolysis, and by upregulating antioxidant defenses to mitigate reactive oxygen species (ROS)-induced oxidative damage. Hypoxia-induced ROS, resulting from mitochondrial dysfunction and enzyme activation, exacerbates genomic instability, tumor aggressiveness, and therapy resistance. Overcoming hypoxia-induced ROS cancer resistance requires a multifaceted approach that targets various aspects of tumor biology. Emerging therapeutic strategies target hypoxia-induced resistance, focusing on hypoxia-inducible factors, ROS levels, and tumor microenvironment subpopulations. Combining innovative therapies with existing treatments holds promise for improving cancer outcomes and overcoming resistance mechanisms.
Collapse
|
23
|
Hindes MT, McElligott AM, Best OG, Ward MP, Selemidis S, Miles MA, Nturubika BD, Gregory PA, Anderson PH, Logan JM, Butler LM, Waugh DJ, O'Leary JJ, Hickey SM, Thurgood LA, Brooks DA. Metabolic reprogramming, malignant transformation and metastasis: Lessons from chronic lymphocytic leukaemia and prostate cancer. Cancer Lett 2025; 611:217441. [PMID: 39755364 DOI: 10.1016/j.canlet.2025.217441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer, crucial for malignant transformation and metastasis. Chronic lymphocytic leukaemia (CLL) and prostate cancer exhibit similar metabolic adaptations, particularly in glucose and lipid metabolism. Understanding this metabolic plasticity is crucial for identifying mechanisms contributing to metastasis. This review considers glucose and lipid metabolism in CLL and prostate cancer, exploring their roles in healthy and malignant states and during disease progression. In CLL, lipid metabolism supports cell survival and migration, with aggressive disease characterised by increased fatty acid oxidation and altered sphingolipids. Richter's transformation and aggressive lymphoma, however, exhibit a metabolic shift towards increased glycolysis. Similarly, prostate cell metabolism is unique, relying on citrate production in the healthy state and undergoing metabolic reprogramming during malignant transformation. Early-stage prostate cancer cells increase lipid synthesis and uptake, and decrease glycolysis, whereas metastatic cells re-adopt glucose metabolism, likely driven by interactions with the tumour microenvironment. Genetic drivers including TP53 and ATM mutations connect metabolic alterations to disease severity in these two malignancies. The bone microenvironment supports the metabolic demands of these malignancies, serving as an initiation niche for CLL and a homing site for prostate cancer metastases. By comparing these malignancies, this review underscores the importance of metabolic plasticity in cancer progression and highlights how CLL and prostate cancer may be models of circulating and solid tumours more broadly. The metabolic phenotypes throughout cancer cell transformation and metastasis, and the microenvironment in which these processes occur, present opportunities for interventions that could disrupt metastatic processes and improve patient outcomes.
Collapse
Affiliation(s)
- Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Anthony M McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Bukuru D Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Solid Tumour Program, Precision Cancer Medicine theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - David J Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
24
|
da Silva Rosa SC, Alizadeh J, Vitorino R, Surendran A, Ravandi A, Kidane B, Ghavami S. A Lipidomics Approach to Determine the Role of Lipids and Its Crosstalk with Autophagy in Lung Cancer Metastasis. Methods Mol Biol 2025; 2879:239-260. [PMID: 38441721 DOI: 10.1007/7651_2024_524] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
Non-small cell lung cancer (NSCLC) is among the most malignant tumors with high propensity for metastasis and is the leading cause of cancer-related death globally. Most patients present with regional and distant metastasis, associated with poor prognosis. Lipids may play an essential role in either activating or inhibiting detachment-induced apoptosis (anoikis), where the latter is a crucial mechanism to prevent metastasis, and it may have a cross-talk with autophagy. Autophagy has been shown to be induced in various human cancer metastasis, modulating tumor cell motility and invasion, cancer cell differentiation, resistance to anoikis, and epithelial to mesenchymal transition. Hence, it may play a crucial role in the transition of benign to malignant phenotypes, the core of metastasis initiation. Here, we provide a method we have established in our laboratory for detecting lipids in attached and detached non-small lung cancer cells and show how to analyze lipidomics data to find its correlation with autophagy-related pathways.
Collapse
Affiliation(s)
- Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rui Vitorino
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, Aveiro, Portugal
| | - Arun Surendran
- Mass Spectrometry Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Biniam Kidane
- Department of Surgery, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
25
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
26
|
Komissarov AA, Zinovyeva MV, Sass AV, Vinogradova TV, Koshechkin SI, Demkin VV, Zborovskaya IB, Kostrov SV, Demidyuk IV. Alterations in the Expression of Proprotein Convertase Genes in Human Esophagus Squamous Cell Carcinomas. Acta Naturae 2025; 17:64-70. [PMID: 40264581 PMCID: PMC12011192 DOI: 10.32607/actanaturae.27437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 03/16/2025] [Indexed: 04/24/2025] Open
Abstract
Proprotein convertases (PCs) constitute an enzyme family that includes nine highly specific human subtilisin-like serine proteases. It is known that the PCs mRNA levels vary in tumors, and that these proteases are involved in carcinogenesis. Thus, PCs may be considered as potential markers for typing and predicting the course of the disease, as well as potential targets for therapy. We used quantitative real-time PCR to evaluate the expression levels of PC genes in the paired samples of tumor and adjacent normal tissues derived from 19 patients with esophageal squamous cell carcinomas. We observed a significant enrichment of PCSK6, PCSK9, MBTPS1, and FURIN mRNAs in the tumor tissue, which may be indication of the involvement of these PCs in the development and progression of esophageal cancers. Additionally, cluster analysis of PC expression alteration patterns in tumor compared to normal adjacent tissues (esophageal and previously analyzed lung tissue samples) revealed a limited set of scenarios for the changes in PC expression. These scenarios are implemented during malignant transformation of lung and esophagus cells, as well as, probably, the cells of other organs. These findings indicate that PC genes may be important markers of human cancers.
Collapse
Affiliation(s)
- A. A. Komissarov
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| | - M. V. Zinovyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - A. V. Sass
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - T. V. Vinogradova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russian Federation
| | - S. I. Koshechkin
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| | - V. V. Demkin
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| | - I. B. Zborovskaya
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russian Federation
| | - S. V. Kostrov
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| | - I. V. Demidyuk
- National Research Centre “Kurchatov Institute”, Moscow, 123182 Russian Federation
| |
Collapse
|
27
|
Zwara A, Hellmann A, Czapiewska M, Korczynska J, Sztendel A, Mika A. The influence of cancer on the reprogramming of lipid metabolism in healthy thyroid tissues of patients with papillary thyroid carcinoma. Endocrine 2025; 87:273-280. [PMID: 39145825 PMCID: PMC11739254 DOI: 10.1007/s12020-024-03993-z] [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: 06/19/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Over the years we observed changes in the metabolism of glucose, amino acids, fatty acids (FA) and nucleotides in cancer cells in order to maintain their viability and proliferate. Moreover, as the latest data show, cancer also forces a complete change in the behavior of other tissues. For instance, fat-filled adipocytes are often found in the vicinity of invasive solid human tumors. We investigated the effects of papillary thyroid carcinoma (PTC) on the lipid metabolism of healthy tissue distant from the tumor. METHOD Thyroid tissue was collected from female patients immediately after surgical removal of the entire thyroid gland. Blood samples were collected from PTC patients and healthy volunteers. Real-time PCR assays were performed to analyze the expression of lipogenic genes and a broad panel of FA was determined using the gas chromatography-mass spectrometry method. RESULTS The concentration of lipids was higher in paratumor tissue than in healthy thyroid tissue (p = 0.005). The lipogenic genes tested were significantly increased in paratumor tissue compared to healthy tissue, especially enzymes related to the synthesis of very long-chain saturated and polyunsaturated FAs (VLCSFAs and PUFAs, respectively) (p < 0.001). The FA profile also showed increased levels of C22-C26, VLCSFAs and almost all PUFAs in paratumor tissue (p < 0.05). CONCLUSION Our study suggests that a restructuring of lipid metabolism occurs in the adjacent healthy thyroid gland and that the metabolism of VLCSFAs and PUFAs is higher in the paratumor tissue than in the distant tissue of the healthy thyroid gland.
Collapse
Affiliation(s)
- Agata Zwara
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland.
| | - Monika Czapiewska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Justyna Korczynska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Alicja Sztendel
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Adriana Mika
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
28
|
Nishizawa H, Funasaki S, Ma W, Kubota Y, Watanabe K, Arima Y, Kuroda S, Ito T, Furuya M, Motoshima T, Nishiyama A, Mehanna S, Satou Y, Hasumi H, Jikuya R, Makiyama K, Tamura T, Oike Y, Tanaka Y, Suda T, Schmidt LS, Linehan WM, Baba M, Kamba T. HIF1α Plays a Crucial Role in the Development of TFE3-Rearranged Renal Cell Carcinoma by Orchestrating a Metabolic Shift Toward Fatty Acid Synthesis. Genes Cells 2025; 30:e13195. [PMID: 39807625 PMCID: PMC11729263 DOI: 10.1111/gtc.13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Tumor development often requires cellular adaptation to a unique, high metabolic state; however, the molecular mechanisms that drive such metabolic changes in TFE3-rearranged renal cell carcinoma (TFE3-RCC) remain poorly understood. TFE3-RCC, a rare subtype of RCC, is defined by the formation of chimeric proteins involving the transcription factor TFE3. In this study, we analyzed cell lines and genetically engineered mice, demonstrating that the expression of the chimeric protein PRCC-TFE3 induced a hypoxia-related signature by transcriptionally upregulating HIF1α and HIF2α. The upregulation of HIF1α by PRCC-TFE3 led to increased cellular ATP production by enhancing glycolysis, which also supplied substrates for the TCA cycle while maintaining mitochondrial oxidative phosphorylation. We crossed TFE3-RCC mouse models with Hif1α and/or Hif2α knockout mice and found that Hif1α, rather than Hif2α, is essential for tumor development in vivo. RNA-seq and metabolomic analyses of the kidney tissues from these mice revealed that ketone body production is inversely correlated with tumor development, whereas de novo lipid synthesis is upregulated through the HIF1α/SREBP1-dependent mechanism in TFE3-RCC. Our data suggest that the coordinated metabolic shift via the PRCC-TFE3/HIF1α/SREBP1 axis is a key mechanism by which PRCC-TFE3 enhances cancer cell metabolism, promoting tumor development in TFE3-RCC.
Collapse
Grants
- JP21K19721 Japan Society for the Promotion of Science
- HHSN261201500003C NCI NIH HHS
- JP24K09315 Japan Society for the Promotion of Science
- JP 24K02578 Japan Society for the Promotion of Science
- JPMXP0618217493 Ministry of Education, Culture, Sports, Science and Technology
- JP20K09560 Japan Society for the Promotion of Science
- JPMXP0622717006 Ministry of Education, Culture, Sports, Science and Technology
- JP21K09374 Japan Society for the Promotion of Science
- JP23K24474 Japan Society for the Promotion of Science
- JP21K06000 Japan Society for the Promotion of Science
- HHSN261201500003I NCI NIH HHS
- JP23K27589 Japan Society for the Promotion of Science
- JPMXP0723833149 Ministry of Education, Culture, Sports, Science and Technology
- Japan Society for the Promotion of Science
- Ministry of Education, Culture, Sports, Science and Technology
- National Cancer Institute
Collapse
Affiliation(s)
- Hidekazu Nishizawa
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Shintaro Funasaki
- Divison of Molecular and Vascular Biology, IRDAKumamoto UniversityKumamotoJapan
| | - Wenjuan Ma
- Cambridge Stem Cell Institute, University of CambridgeCambridgeUK
| | - Yoshiaki Kubota
- Department of AnatomyInstitute for Advanced Medical Research and Keio University School of MedicineTokyoJapan
| | | | - Yuichiro Arima
- Developmental Cardiology Laboratory, International Research Center for Medical Science (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Shoichiro Kuroda
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Takaaki Ito
- Department of Medical TechnologyKumamoto Health Science University Faculty of Health SciencesKumamotoJapan
| | - Mitsuko Furuya
- Department of Surgical PathologyHokkaido University HospitalSapporoJapan
| | - Takanobu Motoshima
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Akira Nishiyama
- Department of ImmunologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Sally Mehanna
- Biotechnology Department, Faculty of Nanotechnology for Postgraduate Studies, Cairo UniversityAd DoqiEgypt
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Hisashi Hasumi
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Ryosuke Jikuya
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Kazuhide Makiyama
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Tomohiko Tamura
- Department of ImmunologyYokohama City University Graduate School of MedicineKanagawaJapan
- Advanced Medical Research CenterYokohama City UniversityKanagawaJapan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Toshio Suda
- Laboratory of Stem Cell Regulation, International Research Center for Medical Science (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Laura S. Schmidt
- Urologic Oncology BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Basic Science Program, Frederick National Laboratory for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - W. Marston Linehan
- Urologic Oncology BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Masaya Baba
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
29
|
Fiorica F, Tebano U, Napoli G, Franceschetto A, Muraro M, Giorgi C, Pinton P. Metabolic-Modulating Effects of Radiation: Undetectable Yet Deadly-A Review on Radiotherapy. Cancers (Basel) 2024; 17:54. [PMID: 39796683 PMCID: PMC11719003 DOI: 10.3390/cancers17010054] [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: 11/27/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
From a cancer-centric perspective, radiotherapy has been primarily viewed as a localised treatment modality, targeting cancer tissues with ionising radiation to induce DNA damage and cell death [...].
Collapse
Affiliation(s)
- Francesco Fiorica
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37122 Verona, Italy; (U.T.); (G.N.); (A.F.); (M.M.)
| | - Umberto Tebano
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37122 Verona, Italy; (U.T.); (G.N.); (A.F.); (M.M.)
| | - Giuseppe Napoli
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37122 Verona, Italy; (U.T.); (G.N.); (A.F.); (M.M.)
| | - Antonella Franceschetto
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37122 Verona, Italy; (U.T.); (G.N.); (A.F.); (M.M.)
| | - Marco Muraro
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37122 Verona, Italy; (U.T.); (G.N.); (A.F.); (M.M.)
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (P.P.)
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (P.P.)
| |
Collapse
|
30
|
Kaimuangpak K, Srisongkram T, Lehtonen M, Rautio J, Weerapreeyakul N. The metabolic response of HepG2 cells to extracellular vesicles derived from Raphanus sativus L. var. caudatus Alef microgreens probed by chemometrics-assisted LC-MS/MS analysis. Food Chem 2024; 461:140833. [PMID: 39151349 DOI: 10.1016/j.foodchem.2024.140833] [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: 05/23/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Extracellular vesicles (EVs) derived from Thai rat-tailed radish (Raphanus sativus L. var. caudatus Alef) microgreens were previously reported as novel bioactive bioparticles against cancer. This study aimed to investigate the metabolic disruption associated with the antiproliferative effect against HepG2 liver cancer cells, a representative of metabolizing cells and tissue. In this study, the neutral red uptake assay was performed to screen for the antiproliferative effect and determine the cytotoxic concentrations of EVs against HepG2 cells. An untargeted approach to cellular metabolomics was conducted using liquid chromatography coupled with the high-resolution mass spectrometry system with multivariate and univariate analyses to determine the metabolic changes of HepG2 liver cancer cells after EV treatment. EVs showed an antiproliferative effect in HepG2 cells with a half-maximal inhibitory concentration (IC50) of 685.5 ± 26.4 and 139.7 ± 4.2 μg/ml at 24 and 48 h, respectively. In the metabolomics study, 163 metabolites were annotated, with 61 significantly altered metabolites. Among these significant metabolites, 18 were related to glycerophospholipid metabolism. Phosphatidylcholine-the important lipid building blocks for cell membranes, lipid mediators for cell proliferation, and immunosuppressive signaling-was mainly decreased by EV treatment. The alteration of cellular phospholipids in cancer was discussed. This finding suggested the possible mechanism of anticancer action of EVs by disrupting phospholipid metabolism and survival signaling in cancer cells. Further studies should be made to confirm EVs' potential as single and combination therapy in vivo to reduce cancer resistance. This may close the gap between in vitro study and clinical setting.
Collapse
Affiliation(s)
- Karnchanok Kaimuangpak
- Graduate School (in the program of Research and Development in Pharmaceuticals), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.
| | - Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand; Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand.
| | - Marko Lehtonen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland.
| | - Jarkko Rautio
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland.
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand; Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
31
|
Yue N, Jin Q, Li C, Zhang L, Cao J, Wu C. CD36: a promising therapeutic target in hematologic tumors. Leuk Lymphoma 2024; 65:1749-1765. [PMID: 38982639 DOI: 10.1080/10428194.2024.2376178] [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: 05/20/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
Cluster of differentiation 36 (CD36) is a multiligand receptor with important roles in lipid metabolism, angiogenesis and innate immunity, and its diverse effects may depend on the binding of specific ligands in different contexts. CD36 is expressed not only on immune cells in the tumor microenvironment (TME) but also on some hematopoietic cells. CD36 is associated with the growth, metastasis and drug resistance in some hematologic tumors, such as leukemia, lymphoma and myelodysplastic syndrome. Currently, some targeted therapeutic agents against CD36 have been developed, such as anti-CD36 antibodies, CD36 antagonists (small molecules) and CD36 expression inhibitors. This paper not only innovatively addresses the role of CD36 in some hematopoietic cells, such as erythrocytes, hematopoietic stem cells and platelets, but also pays special attention to the role of CD36 in the development of hematologic tumors, and suggests that CD36 may be a potential cancer therapeutic target in hematologic tumors.
Collapse
Affiliation(s)
- Ningning Yue
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiqi Jin
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Cuicui Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Litian Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiajia Cao
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chongyang Wu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
32
|
Tsai CC, Wang CY, Chang HH, Chang PTS, Chang CH, Chu TY, Hsu PC, Kuo CY. Diagnostics and Therapy for Malignant Tumors. Biomedicines 2024; 12:2659. [PMID: 39767566 PMCID: PMC11726849 DOI: 10.3390/biomedicines12122659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Malignant tumors remain one of the most significant global health challenges and contribute to high mortality rates across various cancer types. The complex nature of these tumors requires multifaceted diagnostic and therapeutic approaches. This review explores current advancements in diagnostic methods, including molecular imaging, biomarkers, and liquid biopsies. It also delves into the evolution of therapeutic strategies, including surgery, chemotherapy, radiation therapy, and novel targeted therapies such as immunotherapy and gene therapy. Although significant progress has been made in the understanding of cancer biology, the future of oncology lies in the integration of precision medicine, improved diagnostic tools, and personalized therapeutic approaches that address tumor heterogeneity. This review aims to provide a comprehensive overview of the current state of cancer diagnostics and treatments while highlighting emerging trends and challenges that lie ahead.
Collapse
Affiliation(s)
- Chung-Che Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (C.-C.T.); (C.-H.C.); (T.Y.C.)
| | - Chun-Yu Wang
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Hsu-Hung Chang
- Division of Nephrology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 221, Taiwan;
| | | | - Chuan-Hsin Chang
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (C.-C.T.); (C.-H.C.); (T.Y.C.)
| | - Tin Yi Chu
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (C.-C.T.); (C.-H.C.); (T.Y.C.)
| | - Po-Chih Hsu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- Institute of Oral Medicine and Materials, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan; (C.-C.T.); (C.-H.C.); (T.Y.C.)
| |
Collapse
|
33
|
Jardanowska-Kotuniak M, Dramiński M, Własnowolski M, Łapiński M, Sengupta K, Agarwal A, Filip A, Ghosh N, Pancaldi V, Grynberg M, Saha I, Plewczynski D, Dąbrowski MJ. Unveiling epigenetic regulatory elements associated with breast cancer development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623187. [PMID: 39605637 PMCID: PMC11601335 DOI: 10.1101/2024.11.12.623187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Breast cancer is the most common cancer in women and the 2nd most common cancer worldwide, yearly impacting over 2 million females and causing 650 thousand deaths. It has been widely studied, but its epigenetic variation is not entirely unveiled. We aimed to identify epigenetic mechanisms impacting the expression of breast cancer related genes to detect new potential biomarkers and therapeutic targets. We considered The Cancer Genome Atlas database with over 800 samples and several omics datasets such as mRNA, miRNA, DNA methylation, which we used to select 2701 features that were statistically significant to differ between cancer and control samples using the Monte Carlo Feature Selection and Interdependency Discovery algorithm, from an initial total of 417,486. Their biological impact on cancerogenesis was confirmed using: statistical analysis, natural language processing, linear and machine learning models as well as: transcription factors identification, drugs and 3D chromatin structure analyses. Classification of cancer vs control samples on the selected features returned high classification weighted Accuracy from 0.91 to 0.98 depending on feature-type: mRNA, miRNA, DNA methylation, and classification algorithm. In general, cancer samples showed lower expression of differentially expressed genes and increased β-values of differentially methylated sites. We identified mRNAs whose expression is well explained by miRNA expression and differentially methylated sites β-values. We recognized differentially methylated sites possibly affecting NRF1 and MXI1 transcription factors binding, causing a disturbance in NKAPL and PITX1 expression, respectively. Our 3D models showed more loosely packed chromatin in cancer. This study successfully points out numerous possible regulatory dependencies.
Collapse
Affiliation(s)
- Marta Jardanowska-Kotuniak
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Dramiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michał Własnowolski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Marcin Łapiński
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Kaustav Sengupta
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Abhishek Agarwal
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Adam Filip
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Nimisha Ghosh
- Department of Computer Science and Information Technology, Institute of Technical Education and Research, Siksha O Anusandhan University, Bhubaneswar, Odisha, 751030, India
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Indrajit Saha
- Department of Computer Science and Engineering, National Institute of Technical Teachers’ Training and Research, Kolkata 700106, India
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Michał J. Dąbrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
34
|
Zhu Z, Jiang W, Zhou J, Maldeney A, Liang J, Yang J, Luo W. The Combined Inhibition of SREBP and mTORC1 Signaling Synergistically Inhibits B-Cell Lymphoma. Cancer Med 2024; 13:e70342. [PMID: 39501600 PMCID: PMC11538279 DOI: 10.1002/cam4.70342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/20/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The sterol regulatory element-binding protein (SREBP) pathway is essential for maintaining sterol homeostasis during B cell activation and germinal center B cell proliferation. However, its potential as a therapeutic target to treat B-cell lymphoma remains unclear. METHODS We examined SREBP protein expression in human B-cell lymphoma samples using immunohistochemistry. Additionally, we conducted in vitro studies using SREBP signaling inhibitors in combination with rapamycin to assess their effects on cell proliferation and lipid metabolism in B-cell lymphoma cells. RESULTS Our analysis revealed high levels of SREBP2 protein expression in human B-cell lymphoma samples. Inhibiting SREBP signaling or its downstream target HMG-CoA reductase (HMGCR) with Fatostatin or Simvastatin effectively suppressed B-cell lymphoma cell proliferation. However, B-cell lymphoma cells responded to statin treatment by activating the mTORC1-pS6 pathway, suggesting a compensatory mechanism to overcome statin-induced cell cycle arrest. Combining low-dose statin treatment with the mTOR inhibitor rapamycin produced a synergistic effect, significantly inhibiting B-cell lymphoma proliferation, cell cycle progression, and lipid raft formation. CONCLUSIONS These results highlight the potential of a combined therapeutic approach targeting both SREBP and mTORC1 as a novel strategy for treating B-cell lymphoma.
Collapse
Affiliation(s)
- Zhenhan Zhu
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Wenxia Jiang
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jiehao Zhou
- Department of Laboratory Medicine and PathologyMayo Clinic ArizonaPhoenixArizonaUSA
| | - Alexander Robert Maldeney
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jingru Liang
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jing Yang
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Wei Luo
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana University Cooperative Center of Excellence in Hematology (CCEH)Indiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
35
|
Cheng X, Sun G, Meng L, Liu Y, Wen J, Zhao X, Cai W, Xin H, Liu Y, Hao C. Exploring the Molecular Mechanisms of Herbs in the Treatment of Hyperlipidemia Based on Network Pharmacology and Molecular Docking. J Med Food 2024; 27:1092-1105. [PMID: 39149800 DOI: 10.1089/jmf.2024.k.0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Affiliation(s)
- Xiao Cheng
- School of Medicine, Linyi University, Linyi, China
| | - Geng Sun
- School of Chinese Medicine, Bozhou University, Bozhou, China
| | - Li Meng
- School of Medicine, Linyi University, Linyi, China
| | - Yueli Liu
- School of Medicine, Linyi University, Linyi, China
| | - Jiangnan Wen
- School of Medicine, Linyi University, Linyi, China
| | - Xiaoli Zhao
- School of Medicine, Linyi University, Linyi, China
| | - Wenhui Cai
- School of Medicine, Linyi University, Linyi, China
| | - Huawei Xin
- School of Medicine, Linyi University, Linyi, China
| | - Yu Liu
- School of Chinese Medicine, Bozhou University, Bozhou, China
| | | |
Collapse
|
36
|
Ramarajan MG, Parthasarathy KTS, Gaikwad KB, Joshi N, Garapati K, Kandasamy RK, Sharma J, Pandey A. Alterations in Hurler-Scheie Syndrome Revealed by Mass Spectrometry-Based Proteomics and Phosphoproteomics Analysis. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:548-562. [PMID: 39469785 DOI: 10.1089/omi.2024.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Hurler-Scheie syndrome (MPS IH/S), also known as mucopolysaccharidosis type I-H/S (MPS IH/S), is a lysosomal storage disorder caused by deficiency of the enzyme alpha-L-iduronidase (IDUA) leading to the accumulation of glycosaminoglycans (GAGs) in various tissues, resulting in a wide range of symptoms affecting different organ systems. Postgenomic omics technologies offer the promise to understand the changes in proteome, phosphoproteome, and phosphorylation-based signaling in MPS IH/S. Accordingly, we report here a large dataset and the proteomic and phosphoproteomic analyses of fibroblasts derived from patients with MPS IH/S (n = 8) and healthy individuals (n = 8). We found that protein levels of key lysosomal enzymes such as cathepsin D, prosaposin, arylsulfatases (arylsulfatase A and arylsulfatase B), and IDUA were downregulated. We identified 16,693 unique phosphopeptides, corresponding to 4,605 proteins, in patients with MPS IH/S. We found that proteins related to the cell cycle, mitotic spindle assembly, apoptosis, and cytoskeletal organization were differentially phosphorylated in MPS IH/S. We identified 12 kinases that were differentially phosphorylated, including hyperphosphorylation of cyclin-dependent kinases 1 and 2, hypophosphorylation of myosin light chain kinase, and calcium/calmodulin-dependent protein kinases. Taken together, the findings of the present study indicate significant alterations in proteins involved in cytoskeletal changes, cellular dysfunction, and apoptosis. These new observations significantly contribute to the current understanding of the pathophysiology of MPS IH/S specifically, and the molecular mechanisms involved in the storage of GAGs in MPS more generally. Further translational clinical omics studies are called for to pave the way for diagnostics and therapeutics innovation for patients with MPS IH/S.
Collapse
Affiliation(s)
- Madan Gopal Ramarajan
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - K T Shreya Parthasarathy
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Kiran Bharat Gaikwad
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Neha Joshi
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kishore Garapati
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jyoti Sharma
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
37
|
El-Masry TA, El-Nagar MMF, Oriquat GA, Alotaibi BS, Saad HM, El Zahaby EI, Ibrahim HA. Therapeutic efficiency of Tamoxifen/Orlistat nanocrystals against solid ehrlich carcinoma via targeting TXNIP/HIF1-α/MMP-9/P27 and BAX/Bcl2/P53 signaling pathways. Biomed Pharmacother 2024; 180:117429. [PMID: 39293373 DOI: 10.1016/j.biopha.2024.117429] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orlistat (Orli) is an anti-obesity medication that has been approved by the US Food and Drug Administration. It has relatively limited oral bioavailability with promising inhibitory effects on cell proliferation as well as reducing the growth of tumors. AIMS This investigation was done to evaluate the potential protective effect of Tamoxifen/Orlistat nanocrystals alone or in combination against Solid Ehrlich Carcinoma (SEC) and to clarify the possible underlying influences. MATERIALS AND METHODS The liquid antisolvent precipitation technique (bottom-up technology) was utilized to manufacture Orlistat Nanocrystals. To explore potential causes for the anti-tumor action, female Swiss Albino mice bearing SEC were randomly assigned into five equal groups (n = 6). Group 1: Tumor control group, group 2: Tam group: tamoxifen (0.01 g/kg, IP), group 3: Free-Orli group: orlistat (0.24 g/kg, IP), group 4: Nano-Orli: orlistat nanocrystals (0.24 g/kg, IP), group 5: Tam-Nano-Orli: Both doses of Tam and Nano-Orli. All treatments were administered for 16 days. KEY FINDINGS The untreated mice showed development in the tumor volume and weight. As well as histopathology results from these mice revealed many tumor large cells as well as solid sheets of malignant cells. Also, untreated mice showed raised VEGF and TGF-1beta content. Moreover, results of gene expression in the SEC-bearing mice noted upregulation in HIF-1α, MMP-9, Bcl-2, and P27 gene expression and downregulation of TXNIP, BAX, and P53 gene expression. On the other hand, administrated TAM, Free-Orli, Nano-Orli, and a combination of Tam-Nano-Orli distinctly suppressed the tumor effects on estimated parameters with special reference to Tam-Nano-Orli. SIGNIFICANCE The developed Tamoxifen/Orlistat nanocrystals combination could be considered a promising approach to augment antitumor effects.
Collapse
Affiliation(s)
- Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Maysa M F El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ghaleb Ali Oriquat
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Cairo 51511, Egypt.
| | - Enas I El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
38
|
Wang J, Wang H, Zhou W, Luo X, Wang H, Meng Q, Chen J, Chen X, Liu Y, Chan DW, Ju Z, Song Z. MOGAT3-mediated DAG accumulation drives acquired resistance to anti-BRAF/anti-EGFR therapy in BRAFV600E-mutant metastatic colorectal cancer. J Clin Invest 2024; 134:e182217. [PMID: 39436710 PMCID: PMC11645146 DOI: 10.1172/jci182217] [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: 04/22/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
BRAFV600E-mutant metastatic colorectal cancer (mCRC) is associated with poor prognosis. The combination of anti-BRAF/anti-EGFR (encorafenib/cetuximab) treatment for patients with BRAFV600E-mutant mCRC improves clinical benefits; unfortunately, inevitable acquired resistance limits the treatment outcome, and the mechanism has not been validated. Here, we discovered that monoacylglycerol O-acyltransferase 3-mediated (MOGAT3-mediated) diacylglycerol (DAG) accumulation contributed to acquired resistance to encorafenib/cetuximab by dissecting a BRAFV600E-mutant mCRC patient-derived xenograft (PDX) model exposed to encorafenib/cetuximab administration. Mechanistically, the upregulated MOGAT3 promoted DAG synthesis and reduced fatty acid oxidation-promoting DAG accumulation and activated PKCα/CRAF/MEK/ERK signaling, driving acquired resistance. Resistance-induced hypoxia promoted MOGAT3 transcriptional elevation; simultaneously, MOGAT3-mediated DAG accumulation increased HIF1A expression at the translation level through PKCα/CRAF/eIF4E activation, strengthening the resistance status. Intriguingly, reducing intratumoral DAG with fenofibrate or PF-06471553 restored the antitumor efficacy of encorafenib/cetuximab in resistant BRAFV600E-mutant mCRC, which interrupted PKCα/CRAF/MEK/ERK signaling. These findings reveal the critical role of the metabolite DAG as a modulator of encorafenib/cetuximab efficacy in BRAFV600E-mutant mCRC, suggesting that fenofibrate might prove beneficial for resistant BRAFV600E-mutant mCRC patients.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Huogang Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Xin Luo
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huijuan Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Qing Meng
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Jiaxin Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoyu Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Yingqiang Liu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - David W. Chan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
39
|
Kelson CO, Tessmann JW, Geisen ME, He D, Wang C, Gao T, Evers BM, Zaytseva YY. Upregulation of Fatty Acid Synthase Increases Activity of β-Catenin and Expression of NOTUM to Enhance Stem-like Properties of Colorectal Cancer Cells. Cells 2024; 13:1663. [PMID: 39404424 PMCID: PMC11475157 DOI: 10.3390/cells13191663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Dysregulated fatty acid metabolism is an attractive therapeutic target for colorectal cancer (CRC). We previously reported that fatty acid synthase (FASN), a key enzyme of de novo synthesis, promotes the initiation and progression of CRC. However, the mechanisms of how upregulation of FASN promotes the initiation and progression of CRC are not completely understood. Here, using Apc/VillinCre and ApcMin mouse models, we show that upregulation of FASN is associated with an increase in activity of β-catenin and expression of multiple stem cell markers, including Notum. Genetic and pharmacological downregulation of FASN in mouse adenoma organoids decreases the activation of β-catenin and expression of Notum and significantly inhibits organoid formation and growth. Consistently, we demonstrate that NOTUM is highly expressed in human CRC and its expression positively correlates with the expression of FASN in tumor tissues. Utilizing overexpression and shRNA-mediated knockdown of FASN, we demonstrate that upregulation of FASN increases β-catenin transcriptional activity, NOTUM expression and secretion, and enhances stem-like properties of human CRC cells. Pharmacological inhibition of NOTUM decreases adenoma organoids growth and proliferation of cancer cells. In summary, upregulation of FASN enhances β-catenin signaling, increases NOTUM expression and stem-like properties of CRC cells, thus suggesting that targeting FASN upstream of the β-catenin/NOTUM axis may be an effective preventative therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Courtney O. Kelson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (C.O.K.); (J.W.T.); (M.E.G.)
| | - Josiane Weber Tessmann
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (C.O.K.); (J.W.T.); (M.E.G.)
| | - Mariah E. Geisen
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (C.O.K.); (J.W.T.); (M.E.G.)
| | - Daheng He
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (D.H.); (C.W.)
| | - Chi Wang
- Biostatistics and Bioinformatics Shared Resource Facility, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (D.H.); (C.W.)
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (T.G.); (B.M.E.)
| | - B. Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (T.G.); (B.M.E.)
| | - Yekaterina Y. Zaytseva
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA; (C.O.K.); (J.W.T.); (M.E.G.)
| |
Collapse
|
40
|
Zhu W, Lusk JA, Pascua V, Djukovic D, Raftery D. Combination of low glucose and SCD1 inhibition impairs cancer metabolic plasticity and growth in MCF-7 cancer cells: a comprehensive metabolomic and lipidomic analysis. Metabolomics 2024; 20:112. [PMID: 39369160 DOI: 10.1007/s11306-024-02179-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Cancer cells exhibit remarkable metabolic plasticity, enabling them to adapt to fluctuating nutrient conditions. This study investigates the impact of a combination of low glucose levels and inhibition of stearoyl-CoA desaturase 1 (SCD1) using A939572 on cancer metabolic plasticity and growth. METHODS A comprehensive metabolomic and lipidomic analysis was conducted to unravel the intricate changes in cellular metabolites and lipids. MCF-7 cells were subjected to low glucose conditions, and SCD1 was inhibited using A939572. The resulting alterations in metabolic pathways and lipid profiles were explored to elucidate the synergistic effects on cancer cell physiology. RESULTS The combination of low glucose and A939572-induced SCD1 inhibition significantly impaired cancer cell metabolic plasticity. Metabolomic analysis highlighted shifts in key glycolytic and amino acid pathways, indicating the cells' struggle to adapt to restricted glucose availability. Lipidomic profiling revealed alterations in lipid composition, implying disruptions in membrane integrity and signaling cascades. CONCLUSION Our findings underscore the critical roles of glucose availability and SCD1 activity in sustaining cancer metabolic plasticity and growth. Simultaneously targeting these pathways emerges as a promising strategy to impede cancer progression. The comprehensive metabolomic and lipidomic analysis provides a detailed roadmap of molecular alterations induced by this combination treatment, that may help identify potential therapeutic targets.
Collapse
Affiliation(s)
- Wentao Zhu
- Northwest Metabolomics Research Center, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - John A Lusk
- Northwest Metabolomics Research Center, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Vadim Pascua
- Northwest Metabolomics Research Center, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Seattle, WA, USA.
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA.
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| |
Collapse
|
41
|
Güleç Taşkıran AE, Karaoğlu DA, Eylem CC, Ermiş Ç, Güderer İ, Nemutlu E, Demirkol Canlı S, Banerjee S. Glutamine withdrawal leads to the preferential activation of lipid metabolism in metastatic colorectal cancer. Transl Oncol 2024; 48:102078. [PMID: 39111172 PMCID: PMC11362781 DOI: 10.1016/j.tranon.2024.102078] [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: 03/31/2024] [Revised: 07/23/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
INTRODUCTION Glutamine is a non-essential amino acid that is critical for cell growth. However, the differential metabolism of l-glutamine in metastatic versus primary colorectal cancer (CRC) has not been evaluated adequately. MATERIALS AND METHODS Differential expression of glutamine-related genes was determined in primary versus metastatic CRC. Univariate Cox regression and hierarchical clustering were used to generate a gene signature for prognostication. Untargeted metabolomics and 18O based fluxomics were used to identify differential metabolite levels and energy turnover in the paired primary (SW480) and metastatic (SW620) CRC cells. Western blot and qRT-PCR were used to validate differential gene expression. Subcellular localization of E-cadherin was determined by immunocytochemistry. Lipid droplets were visualized with Nile Red. RESULTS The GO term "Glutamine metabolism" was significantly enriched in metastatic versus primary tumors. Supporting this, SW620 cells showed decreased membrane localization of E-cadherin and increased motility upon l-Glutamine withdrawal. A glutamine related signature associated with worse prognosis was identified and validated in multiple datasets. A fluxomics assay revealed a slower TCA cycle in SW480 and SW620 cells upon l-Glutamine withdrawal. SW620 cells, however, could maintain high ATP levels. Untargeted metabolomics indicated the preferential metabolism of fatty acids in SW620 but not SW480 cells. Lipids were mainly obtained from the environment rather than by de novo synthesis. CONCLUSIONS Metastatic CRC cells can display aberrant glutamine metabolism. We show for the first time that upon l-glutamine withdrawal, SW620 (but not SW480) cells were metabolically plastic and could metabolize lipids for survival and cellular motility.
Collapse
Affiliation(s)
- Aliye Ezgi Güleç Taşkıran
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye; Department of Molecular Biology and Genetics, Başkent University, Ankara, Türkiye
| | | | - Cemil Can Eylem
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Çağdaş Ermiş
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye
| | - İsmail Güderer
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Seçil Demirkol Canlı
- Division of Tumor Pathology, Department of Clinical Oncology, Cancer Institute, Hacettepe University, Ankara, Türkiye
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye; Cancer Systems Biology (CanSyL), Orta Doğu Teknik Üniversitesi, Ankara, Türkiye.
| |
Collapse
|
42
|
Yang L, Pham K, Xi Y, Jiang S, Robertson KD, Liu C. Acyl-CoA Synthetase Medium-Chain Family Member 5-Mediated Fatty Acid Metabolism Dysregulation Promotes the Progression of Hepatocellular Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1951-1966. [PMID: 39069168 PMCID: PMC11423759 DOI: 10.1016/j.ajpath.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/23/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, with high incidence and mortality worldwide. Despite diagnostic and therapeutic advancements, HCC remains poorly responsive to treatment, with a poor prognosis. Understanding the molecular mechanisms driving HCC is crucial for developing effective therapies. Emerging evidence indicates that dysregulated fatty acid metabolism contributes to HCC. Acyl-CoA medium-chain synthetase 5 (ACSM5), involved in fatty acid metabolism, is down-regulated in HCC; however, its role is not well understood. This study was used to analyze ACSM5 expression in HCC patient samples and cell lines. The newly established ACSM5-overexpressing HCC cell lines, Huh7-ACSM5 and Hepa1-6-ACSM5, were used to investigate the effects and regulatory mechanisms of ACSM5. The results showed that ACSM5 was significantly down-regulated in HCC tumor tissues compared with non-tumor tissues. ACSM5 expression was regulated by DNA methylation, with a DNA methyltransferase 1 (DNMT1) inhibitor effectively increasing ACSM5 expression and reducing promoter region methylation. Overexpression of ACSM5 in Huh7 cells reduced fatty acid accumulation, decreased cell proliferation, migration, and invasion in vitro, and inhibited tumor growth in mouse xenografts. Furthermore, ACSM5 overexpression also decreased STAT3 phosphorylation, subsequently affecting downstream cytokine TGFB and FGF12 mRNA levels. These findings suggest that ACSM5 down-regulation contributes to HCC progression, providing insights into its oncogenic role and highlighting its potential as a biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Lei Yang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Kien Pham
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Yibo Xi
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Shaoning Jiang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
43
|
Nambiar SS, Ghosh SS, Saini GK. Gliotoxin triggers cell death through multifaceted targeting of cancer-inducing genes in breast cancer therapy. Comput Biol Chem 2024; 112:108170. [PMID: 39146703 DOI: 10.1016/j.compbiolchem.2024.108170] [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: 06/18/2024] [Revised: 08/03/2024] [Accepted: 08/03/2024] [Indexed: 08/17/2024]
Abstract
Fungal secondary metabolites have a long history of contributing to pharmaceuticals, notably in the development of antibiotics and immunosuppressants. Harnessing their potent bioactivities, these compounds are now being explored for cancer therapy, by targeting and disrupting the genes that induce cancer progression. The current study explores the anticancer potential of gliotoxin, a fungal secondary metabolite, which encompasses a multi-faceted approach integrating computational predictions, molecular dynamics simulations, and comprehensive experimental validations. In-silico studies have identified potential gliotoxin targets, including MAPK1, NFKB1, HIF1A, TDP1, TRIM24, and CTSD which are involved in critical pathways in cancer such as the NF-κB signaling pathway, MAPK/ERK signaling pathway, hypoxia signaling pathway, Wnt/β-catenin pathway, and other essential cellular processes. The gene expression analysis results indicated all the identified targets are overexpressed in various breast cancer subtypes. Subsequent molecular docking and dynamics simulations have revealed stable binding of gliotoxin with TDP1 and HIF1A. Cell viability assays exhibited a dose-dependent decreasing pattern with its remarkable IC50 values of 0.32, 0.14, and 0.53 μM for MDA-MB-231, MDA-MB-468, and MCF-7 cells, respectively. Likewise, in 3D tumor spheroids, gliotoxin exhibited a notable decrease in viability indicating its effectiveness against solid tumors. Furthermore, gene expression studies using Real-time PCR revealed a reduction of expression of cancer-inducing genes, MAPK1, HIF1A, TDP1, and TRIM24 upon gliotoxin treatment. These findings collectively underscore the promising anticancer potential of gliotoxin through multi-targeting cancer-promoting genes, positioning it as a promising therapeutic option for breast cancer.
Collapse
Affiliation(s)
- Sujisha S Nambiar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 39, India
| | - Gurvinder Kaur Saini
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India.
| |
Collapse
|
44
|
Huang Y, Wei H, Feng H, Tian F, Zheng Q, Deng Z. An endogenous oxygen self-supplied nanoplatform with GSH-depleted and NIR-II triggered electron-hole separation for enhanced photocatalytic anti-tumor therapy. Phys Chem Chem Phys 2024; 26:23386-23392. [PMID: 39212464 DOI: 10.1039/d4cp02554g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The use of artificial enzymes and light energy in photocatalytic therapy, a developing drug-free therapeutic approach, can treat malignant tumors in vivo. However, the relatively deficient oxygen concentration in the tumor microenvironment (TME) restrains their further tumor treatment capability. Herein, a novel nanoplatform with Cu7S4@Au nanocatalyst coated by MnO2 was successfully designed. After 1064 nm light irradiation, the designed nanocatalyst can promote the separation of light generated electron-hole pairs, resulting in ROS generation and tumor cell apoptosis. The MnO2 shelled nanoplatform can function as a TME-responsive oxygen self-supplied producer to improve photocatalyst treatment and GSH depletion. In summary, the designed novel nanoplatform shows efficient inhibition of tumor growth via GSH depletion and synergistic photocatalytic therapy, which is of great significance for improving the clinical tumor treatment effect.
Collapse
Affiliation(s)
- Yao Huang
- School of Physics and Electronic-Electrical Engineering, Xiangnan University, Chenzhou 423000, China
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Hanlin Wei
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Hui Feng
- Changsha Environmental Protection College, Hunan Province, Changsha 410082, China
| | - Fengyu Tian
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Qi Zheng
- Key Laboratory for Micro-Nano Optoelectronic Devices of Ministry of Education, School of Physics and Electronics, Hunan University, Changsha, China.
| | - Zhiming Deng
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| |
Collapse
|
45
|
Zhu X, Huang H, Zou M, Luo H, Liu T, Zhu S, Ye B. Identification of circulating metabolites linked to the risk of breast cancer: a mendelian randomization study. Front Pharmacol 2024; 15:1442723. [PMID: 39323635 PMCID: PMC11422656 DOI: 10.3389/fphar.2024.1442723] [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: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Objective This study aimed to investigate potential causal relationships between circulating metabolites and breast cancer risk using Mendelian randomization (MR) analysis. Materials and Methods Summary-level genome-wide association study (GWAS) datasets for 249 circulating metabolites were obtained from the UK Biobank. GWAS datasets for estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancer were acquired from previous studies based on the Combined Oncoarray. Instrumental variables (IVs) were selected from single nucleotide polymorphisms (SNPs) associated with circulating metabolites, and MR analyses were conducted using the inverse-variance weighted (IVW) method as the primary analysis, with additional sensitivity analyses using other MR methods. Odds ratios (OR) and 95% confidence interval (CI) were used to estimate the association of circulating metabolites with breast cancer risk. Results The IVW analysis revealed significant causal relationships between 79 circulating metabolites and ER + breast cancer risk, and 10 metabolites were significantly associated with ER-breast cancer risk. Notably, acetate (OR = 1.12, P = 0.03), HDL cholesterol (OR = 1.09, P < 0.001), ration of omega-6 fatty acids to total fatty acids ratio (OR = 1.09, P = 0.01), and phospholipids in large LDL (OR = 1.09, P < 0.001) were linked to an increased risk of ER + breast cancer, while linoleic acid (OR = 0.91, P < 0.001) monounsaturated fatty acids (OR = 0.91, P < 0.001), and total lipids in LDL (OR = 0.91, P < 0.001) were associated with a decreased risk. In ER-breast cancer, glycine, citrate, HDL cholesterol, cholesteryl esters in HDL, cholesterol to total lipids ratio in very large HDL, and cholesterol in large LDL were associated with an increased risk, while the free cholesterol to total lipids in very large HDL was linked to a decreased risk. Conclusion This MR approach underscores aberrant lipid metabolism as a key process in breast tumorigenesis, and may inform future prevention and treatment strategies. To further elucidate the underlying mechanisms and explore the potential clinical implications, additional research is warranted to validate the observed associations in this study.
Collapse
Affiliation(s)
- Xiaosheng Zhu
- Department of Radiation, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huai Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Mengjie Zou
- Department of Nephrology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Honglin Luo
- Institute of Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Tianqi Liu
- Department of General Surgery, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shaoliang Zhu
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Bin Ye
- Department of Radiation, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
46
|
Pervushin NV, Yapryntseva MA, Panteleev MA, Zhivotovsky B, Kopeina GS. Cisplatin Resistance and Metabolism: Simplification of Complexity. Cancers (Basel) 2024; 16:3082. [PMID: 39272940 PMCID: PMC11394643 DOI: 10.3390/cancers16173082] [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: 07/25/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Cisplatin is one of the most well-known anti-cancer drugs and has demonstrated efficacy against numerous tumor types for many decades. However, a key challenge with cisplatin, as with any chemotherapeutic agent, is the development of resistance with a resultant loss of efficacy. This resistance is often associated with metabolic alterations that allow insensitive cells to divide and survive under treatment. These adaptations could vary greatly among different tumor types and may seem questionable and incomprehensible at first glance. Here we discuss the disturbances in glucose, lipid, and amino acid metabolism in cisplatin-resistant cells as well as the roles of ferroptosis and autophagy in acquiring this type of drug intolerance.
Collapse
Affiliation(s)
- Nikolay V Pervushin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maria A Yapryntseva
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Mikhail A Panteleev
- Department of Medical Physics, Physics Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, P.O. Box 210, 17177 Stockholm, Sweden
| | - Gelina S Kopeina
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
47
|
Maraqah HH, Aboubechara JP, Abu-Asab MS, Lee HS, Aboud O. Excessive lipid production shapes glioma tumor microenvironment. Ultrastruct Pathol 2024; 48:367-377. [PMID: 39157967 PMCID: PMC11495230 DOI: 10.1080/01913123.2024.2392728] [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: 04/18/2024] [Revised: 05/27/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Disrupted lipid metabolism is a characteristic of gliomas. This study utilizes an ultrastructural approach to characterize the prevalence and distribution of lipids within gliomas. This study made use of tissue from IDH1 wild type (IDH1-wt) glioblastoma (n = 18) and IDH1 mutant (IDH1-mt) astrocytoma (n = 12) tumors. We uncover a prevalent and intriguing surplus of lipids. The bulk of the lipids manifested as sizable cytoplasmic inclusions and extracellular deposits in the tumor microenvironment (TME); in some tumors the lipids were stored in the classical membraneless spheroidal lipid droplets (LDs). Frequently, lipids accumulated inside mitochondria, suggesting possible dysfunction of the beta-oxidation pathway. Additionally, the tumor vasculature have lipid deposits in their lumen and vessel walls; this lipid could have shifted in from the tumor microenvironment or have been produced by the vessel-invading tumor cells. Lipid excess in gliomas stems from disrupted beta-oxidation and dysfunctional oxidative phosphorylation pathways. The implications of this lipid-driven environment include structural support for the tumor cells and protection against immune responses, non-lipophilic drugs, and free radicals.
Collapse
Affiliation(s)
- Haitham H Maraqah
- Medicine & Health Science Faculty, School of Meidicine, An-Najah National University, Nablus, Palestine
| | - John Paul Aboubechara
- Department of Neurology, University of California Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA
| | - Mones S Abu-Asab
- Electron Microscopy Lab, Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Han Sung Lee
- Department of Pathology and Laboratory Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Orwa Aboud
- Department of Neurology, University of California Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA
- Department of Neurosurgery, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
48
|
Bhardwaj JK, Siwach A, Sachdeva SN. Metabolomics and cellular altered pathways in cancer biology: A review. J Biochem Mol Toxicol 2024; 38:e23807. [PMID: 39148273 DOI: 10.1002/jbt.23807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Cancer is a deadly disease that affects a cell's metabolism and surrounding tissues. Understanding the fundamental mechanisms of metabolic alterations in cancer cells would assist in developing cancer treatment targets and approaches. From this perspective, metabolomics is a great analytical tool to clarify the mechanisms of cancer therapy as well as a useful tool to investigate cancer from a distinct viewpoint. It is a powerful emerging technology that detects up to thousands of molecules in tissues and biofluids. Like other "-omics" technologies, metabolomics involves the comprehensive investigation of micromolecule metabolites and can reveal important details about the cancer state that is otherwise not apparent. Recent developments in metabolomics technologies have made it possible to investigate cancer metabolism in greater depth and comprehend how cancer cells utilize metabolic pathways to make the amino acids, nucleotides, and lipids required for tumorigenesis. These new technologies have made it possible to learn more about cancer metabolism. Here, we review the cellular and systemic effects of cancer and cancer treatments on metabolism. The current study provides an overview of metabolomics, emphasizing the current technologies and their use in clinical and translational research settings.
Collapse
Affiliation(s)
- Jitender Kumar Bhardwaj
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Anshu Siwach
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, India
| | - Som Nath Sachdeva
- Department of Civil Engineering, National Institute of Technology, Kurukshetra and Kurukshetra University, Kurukshetra, Haryana, India
| |
Collapse
|
49
|
Lin Z, Hua G, Hu X. Lipid metabolism associated crosstalk: the bidirectional interaction between cancer cells and immune/stromal cells within the tumor microenvironment for prognostic insight. Cancer Cell Int 2024; 24:295. [PMID: 39174964 PMCID: PMC11342506 DOI: 10.1186/s12935-024-03481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Cancer is closely related to lipid metabolism, with the tumor microenvironment (TME) containing numerous lipid metabolic interactions. Cancer cells can bidirectionally interact with immune and stromal cells, the major components of the TME. This interaction is primarily mediated by fatty acids (FAs), cholesterol, and phospholipids. These interactions can lead to various physiological changes, including immune suppression, cancer cell proliferation, dissemination, and anti-apoptotic effects on cancer cells. The physiological modulation resulting from this lipid metabolism-associated crosstalk between cancer cells and immune/stromal cells provides valuable insights into cancer prognosis. A comprehensive literature review was conducted to examine the function of the bidirectional lipid metabolism interactions between cancer cells and immune/stromal cells within the TME, particularly how these interactions influence cancer prognosis. A novel autophagy-extracellular vesicle (EV) pathway has been proposed as a mediator of lipid metabolism interactions between cancer cells and immune cells/stromal cells, impacting cancer prognosis. As a result, different forms of lipid metabolism interactions have been described as being linked to cancer prognosis, including those mediated by the autophagy-EV pathway. In conclusion, understanding the bidirectional lipid metabolism interactions between cancer cells and stromal/immune cells in the TME can help develop more advanced prognostic approaches for cancer patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Guanxiang Hua
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Xiaojuan Hu
- Queen Mary College, Nanchang University, Nanchang, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
50
|
Casacuberta-Serra S, González-Larreategui Í, Capitán-Leo D, Soucek L. MYC and KRAS cooperation: from historical challenges to therapeutic opportunities in cancer. Signal Transduct Target Ther 2024; 9:205. [PMID: 39164274 PMCID: PMC11336233 DOI: 10.1038/s41392-024-01907-z] [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: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 08/22/2024] Open
Abstract
RAS and MYC rank amongst the most commonly altered oncogenes in cancer, with RAS being the most frequently mutated and MYC the most amplified. The cooperative interplay between RAS and MYC constitutes a complex and multifaceted phenomenon, profoundly influencing tumor development. Together and individually, these two oncogenes regulate most, if not all, hallmarks of cancer, including cell death escape, replicative immortality, tumor-associated angiogenesis, cell invasion and metastasis, metabolic adaptation, and immune evasion. Due to their frequent alteration and role in tumorigenesis, MYC and RAS emerge as highly appealing targets in cancer therapy. However, due to their complex nature, both oncogenes have been long considered "undruggable" and, until recently, no drugs directly targeting them had reached the clinic. This review aims to shed light on their complex partnership, with special attention to their active collaboration in fostering an immunosuppressive milieu and driving immunotherapeutic resistance in cancer. Within this review, we also present an update on the different inhibitors targeting RAS and MYC currently undergoing clinical trials, along with their clinical outcomes and the different combination strategies being explored to overcome drug resistance. This recent clinical development suggests a paradigm shift in the long-standing belief of RAS and MYC "undruggability", hinting at a new era in their therapeutic targeting.
Collapse
Affiliation(s)
| | - Íñigo González-Larreategui
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Daniel Capitán-Leo
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Laura Soucek
- Peptomyc S.L., Barcelona, Spain.
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
- Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|