1
|
Ghanbarian M, Dolgova N, Vizeacoumar FS, Vizeacoumar FJ, Michel D, El-Aneed A, Dmitriev OY. Metabolic Effects of the Cancer Metastasis Modulator MEMO1. Metabolites 2025; 15:277. [PMID: 40278406 DOI: 10.3390/metabo15040277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Cancer cells often display altered energy metabolism. In particular, expression levels and activity of the tricarboxylic acid cycle (TCA cycle) enzymes may change in cancer, and dysregulation of the TCA cycle is a frequent hallmark of cancer cell metabolism. MEMO1, a modulator of cancer metastasis, has been shown to bind iron and regulate iron homeostasis in the cells. MEMO1 knockout changed mitochondrial morphology and iron content in breast cancer cells. Our previous genome-wide analysis of MEMO1 genetic interactions across multiple cancer cell lines revealed that gene sets involved in mitochondrial respiration and the TCA cycle are enriched among the gain-of-function interaction partners of MEMO1. Based on these findings, we measured the TCA cycle metabolite levels in breast cancer cells with varying levels of MEMO1 expression. Methods: ShRNA knockdown assay was performed to test essentiality of key TCA cycle enzymes. TCA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in MDA-MB-231 (high MEMO1), M67-2 (MEMO1 knockdown), and M67-9 (MEMO1 knockout) cells under iron-depleted, basal iron, and iron-supplemented conditions. Results:ACO2 and OGDH knockdowns inhibit cell proliferation, indicating an essential role of the TCA cycle in MDA-MB-231 metabolism. α-Ketoglutarate and citrate levels exhibited an inverse relationship with MEMO1 expression, increasing significantly in MEMO1 knockout cells regardless of iron availability. In contrast, fumarate, malate, and glutamate levels were elevated in MEMO1 knockout cells specifically under low iron conditions, suggesting an iron-dependent effect. Conclusions: Overall, our results indicate that MEMO1 plays a role in regulating the TCA in cancer cells in an iron-dependent manner.
Collapse
Affiliation(s)
- Marziyeh Ghanbarian
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Natalia Dolgova
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Frederick S Vizeacoumar
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Franco J Vizeacoumar
- Cancer Research Department, Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
- Division of Oncology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Deborah Michel
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anas El-Aneed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Oleg Y Dmitriev
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
2
|
Zhang L, Li Y, Li Y. Circulating citrate as a mediator in the relationship between HMGCR inhibitors and chronic hepatitis B: a Mendelian randomization study. Sci Rep 2025; 15:12768. [PMID: 40229388 PMCID: PMC11997182 DOI: 10.1038/s41598-025-95100-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: 09/30/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Observational studies have found that HMGCR inhibitors can be used to treat chronic viral hepatitis. In this study, to explore the potential mechanism of HMGCR inhibitors in treating Chronic hepatitis B (CHB), two-sample and two-step Mendelian randomization (MR) were used to investigate the causal relationship between HMGCR inhibitors and the mediating role of circulating metabolites. GWAS data of expression quantitative trait loci eQTLs of HMGCR inhibitors, 168 circulating metabolites, CHB, and myocardial infarction were obtained from the IEUOpenGWAS project. Random effects inverse-variance weighted (IVW) was the main causal analysis method, and the MR-Egger regression method was used as a supplementary analysis method. Cochran's Q test and I2 statistic were used to determine the heterogeneity of SNPs. The intercept terms of the MR-Egger method and MR-PRESSO were used for pleiotropy analysis, and leave-one-out was used for sensitivity analysis. Mediation effect analysis was used to evaluate the mediating role of the circulating metabolites. Genetic variations in the drug target genes of HMGCR inhibitors were associated with a reduced risk of chronic hepatitis B and myocardial infarction (P < 0.05). Eight circulating metabolites had a significant causal relationship with HMGCR inhibitors and CHB. After further calculation of the mediation effect, citrate was used as a mediating variable between HMGCR inhibitors and CHB, with a mediation effect of - 0.015 and a mediation ratio of 9.769%. HMGCR inhibitors can significantly reduce the risk of CHB, and the circulating metabolite citrate may mediate this association. However, this study has certain limitations. The short-term effects of HMGCR inhibitors on CHB could not be assessed, and partial overlap between the GWAS data for HMGCR inhibitors and circulating metabolites may introduce bias in estimating causal effects.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Ultrasonography, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China
| | - Yuming Li
- Department of Advanced Mathematics, Basic Department, Jilin College of Commerce and Industry, No. 1666, Kalun Lake Street, Jiutai Economic Development Zone, Changchun, 130021, Jilin, People's Republic of China
| | - Ying Li
- Department of Neonatology, Children's Medical Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
3
|
Wu Z, Yang J, Ma Z, Chen Y, Han M, Wu Q, Hou B, Huang S, Zhang C. Nuclear magnetic resonance-based metabolomics and risk of pancreatic cancer: a prospective analysis in the UK Biobank. J Gastroenterol 2025:10.1007/s00535-025-02237-9. [PMID: 40074913 DOI: 10.1007/s00535-025-02237-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Plasma metabolite levels in patients with pancreatic cancer (PC) have changed, but the relationship between the altered plasma metabolites and the risk for PC occurrence is not fully clear, as well as the predictive value of the specific metabolites. METHODS In this study, we obtained the metabolomics data of 243,145 people from the UK Biobank. An extreme gradient boosting (XGBoost) model, least absolute shrinkage and selection operator (Lasso) regression, and covariate-adjusted Cox proportional hazard regression models were used to evaluate the relationship between metabolites and PC risk. We also evaluated conventional risks, metabolites, and combination models for PC risk by comparing the area under the receiver operating characteristic curve (AUC). RESULTS The average follow-up time was 13.8 (± 2.1) years; 1,026 of 243,145 participants developed PC. Fourteen metabolites were significantly associated with PC, including glucose-related metabolites, lipids, lipoproteins, and amino acids. Increased PC risk was associated with citrate, glucose, and the percentage of triglycerides to total lipids in intermediate-density lipoprotein or small low-density lipoprotein. Glycine, histidine, cholesterol, and cholesterol ester subclasses were associated with lower PC risk. Predicting PC risk improved when the newly identified metabolites were added to conventional PC risk factors (AUC: 0.705 vs 0.711, p = 0.037). The Kaplan-Meier cumulative incidence curves showed that these metabolites were good predictors of PC risk (all log-rank p < 0.05). CONCLUSION We identified novel metabolites that were significantly associated with the occurrence of PC, which may aid in the early diagnosis of PC.
Collapse
Affiliation(s)
- Zelong Wu
- Department of Hepatological Surgery, Maoming People's Hospital, Maoming, 525000, China
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiayu Yang
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangzhou, 510080, China
| | - Zuyi Ma
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100005, China
| | - Yubin Chen
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- South China University of Technology School of Medicine, Guangzhou, 51000, China
| | - Mingqian Han
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qianlong Wu
- Department of General Surgery, Heyuan People's Hospital, Heyuan, 517000, China
| | - Baohua Hou
- Department of Hepatological Surgery, Maoming People's Hospital, Maoming, 525000, China.
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangzhou, 510080, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
| | - Shanzhou Huang
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangzhou, 510080, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
| | - Chuanzhao Zhang
- DepartmentofGeneralSurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangzhou, 510080, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
- South China University of Technology School of Medicine, Guangzhou, 51000, China.
| |
Collapse
|
4
|
Liu Q, Xue Y, Guo J, Tao L, Zhu Y. Citrate: a key signalling molecule and therapeutic target for bone remodeling disorder. Front Endocrinol (Lausanne) 2025; 15:1512398. [PMID: 39886032 PMCID: PMC11779597 DOI: 10.3389/fendo.2024.1512398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025] Open
Abstract
Bone remodeling is a continuous cyclic process that maintains and regulates bone structure and strength. The disturbance of bone remodeling leads to a series of bone metabolic diseases. Recent studies have shown that citrate, an intermediate metabolite of the tricarboxylic acid (TCA) cycle, plays an important role in bone remodeling. But the exact mechanism is still unclear. In this study, we focused on the systemic regulatory mechanism of citrate on bone remodeling, and found that citrate is involved in bone remodeling in multiple ways. The participation of citrate in oxidative phosphorylation (OXPHOS) facilitates the generation of ATP, thereby providing substantial energy for bone formation and resorption. Osteoclast-mediated bone resorption releases citrate from bone mineral salts, which is subsequently released as an energy source to activate the osteogenic differentiation of stem cells. Finally, the differentiated osteoblasts secrete into the bone matrix and participate in bone mineral salts formation. As a substrate of histone acetylation, citrate regulates the expression of genes related to bone formation and bone reabsorption. Citrate is also a key intermediate in the metabolism and synthesis of glucose, fatty acids and amino acids, which are three major nutrients in the organism. Citrate can also be used as a biomarker to monitor bone mass transformation and plays an important role in the diagnosis and therapeutic evaluation of bone remodeling disorders. Citrate imbalance due to citrate transporter could result in the supression of osteoblast/OC function through histone acetylation, thereby contributing to disorders in bone remodeling. Therefore, designing drugs targeting citrate-related proteins to regulate bone citrate content provides a new direction for the drug treatment of diseases related to bone remodeling disorders.
Collapse
Affiliation(s)
| | | | | | - Lin Tao
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Huang Y, He C, Hu Q, Liu Z, Li X, Gao W, Liang X, Chen R, Mao Z, Lin X. Metabolic Atlas of Human Eyelid Infiltrative Basal Cell Carcinoma. Invest Ophthalmol Vis Sci 2025; 66:11. [PMID: 39775699 PMCID: PMC11717128 DOI: 10.1167/iovs.66.1.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Purpose Eyelid infiltrative basal cell carcinoma (iBCC) is the most common malignant tumor affecting the ocular adnexa, but studies on metabolic changes within its microenvironment and heterogeneity at the tumor invasive area are limited. This study aims to analyze metabolic differences among iBCC cell types using single-cell and spatial metabolomics analysis and to examine metabolic environment at the tumor invasive area. Methods Single-cell transcriptomic data of human basal cell carcinoma (BCC) were clustered and visualized using Uniform Manifold Approximation and Projection. Metabolic reprogramming was analyzed with single-cell flux estimation analysis. Spatial metabolomics data were obtained with the Timstof Flex MALDI 2 system, and Bruker software was used for region selection. Results Eight cell types were identified within the iBCC microenvironment. Differences between inflammatory cancer-associated fibroblasts and myofibroblastic cancer-associated fibroblasts were analyzed. Metabolic flux analysis showed increased glycolysis, glutamine, heme, and glutathione fluxes in the iBCC microenvironment. Spatial metabolomics revealed high levels of taurine, deoxy-GMP, O-phosphoethanolamine, and pyrithione. Both tumor and invasive regions had significant upregulation of fatty acid pathways, with marked increases in oleic and arachidonic acids at the invasive area. Specific upregulation of UDP-glucuronic acid and high UDP-glucose 6-dehydrogenase (UGDH) expression in the tumor region suggest UXS1 as a potential therapeutic target for iBCC. Conclusions This study establishes a metabolic microenvironment atlas of iBCC, revealing significant metabolic differences and the dominance of lipid and lysosome metabolism. Potential metabolic markers and characteristic substances in the invasive area offer new insights for immunotherapy and the exploration of BCC's metabolic mechanisms.
Collapse
Affiliation(s)
- Yanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chengjie He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qiuling Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xingyi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Wuyou Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuanwei Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rongxin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhen Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
6
|
Icard P, Prieto M, Coquerel A, Fournel L, Gligorov J, Noel J, Mouren A, Dohan A, Alifano M, Simula L. Why and how citrate may sensitize malignant tumors to immunotherapy. Drug Resist Updat 2025; 78:101177. [PMID: 39612545 DOI: 10.1016/j.drup.2024.101177] [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: 12/19/2023] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Immunotherapy, either alone or in combination with chemotherapy, has demonstrated limited efficacy in a variety of solid cancers. Several factors contribute to explaining primary or secondary resistance. Among them, cancer cells, whose metabolism frequently relies on aerobic glycolysis, promote exhaustion of cytotoxic immune cells by diverting the glucose in the tumor microenvironment (TME) to their own profit, while secreting lactic acid that sustains the oxidative metabolism of immunosuppressive cells. Here, we propose to combine current treatment based on the use of immune checkpoint inhibitors (ICIs) with high doses of sodium citrate (SCT) because citrate inhibits cancer cell metabolism (by targeting both glycolysis and oxidative metabolism) and may active anti-tumor immune response. Indeed, as showed in preclinical studies, SCT reduces cancer cell growth, promoting cell death and chemotherapy effectiveness. Furthermore, since the plasma membrane citrate carrier pmCIC is mainly expressed in cancer cells and low or not expressed in immune and non-transformed cells, we argue that the inhibition of cancer cell metabolism by SCT may increase glucose availability in the TME, thus promoting functionality of anti-tumor immune cells. Concomitantly, the decrease in the amount of lactic acid in the TME may reduce the functionality of immunosuppressive cells. Preclinical studies have shown that SCT can enhance the anti-tumor immune response through an enhancement of T cell infiltration and activation, and a repolarization of macrophages towards a TAM1-like phenotype. Therefore, this simple and cheap strategy may have a major impact to increase the efficacy of current immunotherapies in human solid tumors and we encourage testing it in clinical trials.
Collapse
Affiliation(s)
- Philippe Icard
- INSERM U1086 ANTICIPE, Interdisciplinary Research Unit for Cancers Prevention and Treatment, BioTICLA Laboratory, Université de Caen Normandie, Caen, France; Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France.
| | - Mathilde Prieto
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Antoine Coquerel
- INSERM U1075, COMETE « Mobilités: Attention, Orientation, Chronobiologie », Université Caen, France
| | - Ludovic Fournel
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France; INSERM UMR-S 1007, Cellular Homeostasis and Cancer, Paris-Descartes University, Paris
| | - Joseph Gligorov
- Oncology Department, Tenon Hospital, Pierre et Marie Curie University, Paris
| | - Johanna Noel
- Oncology Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Adrien Mouren
- Département d'Innovation Thérapeutique et d´Essais Précoces (DITEP), Institut Gustave Roussy, Villejuif 94805, France
| | - Anthony Dohan
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris 75014, France; Radiology Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, Paris-Descartes University, Paris, France
| | - Luca Simula
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris 75014, France.
| |
Collapse
|
7
|
Mohd Faizal NF, Vincent-Chong VK, Ramanathan A, Paterson IC, Karen-Ng LP, Zaini ZM. Metabolomic Profiling of Oral Potentially Malignant Disorders and Its Clinical Values. Biomedicines 2024; 12:2899. [PMID: 39767805 PMCID: PMC11726734 DOI: 10.3390/biomedicines12122899] [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: 10/26/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Oral potentially malignant disorders (OPMD) are a group of lesions carrying the risk of developing into cancer. The gold standard to predict which lesions are more likely to undergo malignant transformation is the presence of dysplasia histologically. However, not all dysplastic lesions progress, and non-dysplastic lesions may also undergo malignant transformation. Oral carcinogenesis is a complex molecular process that involves somatic alterations and the deregulation of transcriptions, protein expression, and metabolite levels. Metabolomics, which is the scientific study of metabolites, has emerged as a promising high-throughput approach to investigate the metabolic changes of small molecules in biological pathways. In this review, we summarize the data relating to the metabolomic profiling of OPMDs, which will help elucidate the complex process of oral carcinogenesis. Furthermore, we identify that among all metabolites, citrate, pyruvate, and glutamate may serve as potential biomarkers for oral leukoplakia (OLK). Notably, metformin and gluconate have been shown to target glutamate and citrate, respectively, in cancer cells. Based on these findings, we propose that targeting these metabolites in patients with OPMD could be a promising therapeutic strategy to mitigate OPMD progression and potentially reduce the risk of malignant transformation. We also discuss the limitations and future directions of metabolomics in OPMD. Understanding these important metabolites is crucial for early detection and monitoring of oral cancer progression.
Collapse
Affiliation(s)
- Nur Fatinazwa Mohd Faizal
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (N.F.M.F.); (A.R.)
| | - Vui King Vincent-Chong
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Anand Ramanathan
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (N.F.M.F.); (A.R.)
- Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Ian C. Paterson
- Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Lee Peng Karen-Ng
- Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Zuraiza Mohamad Zaini
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (N.F.M.F.); (A.R.)
- Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| |
Collapse
|
8
|
Wu H, Li Z, Yang L, He L, Liu H, Yang S, Xu Q, Li Y, Li W, Li Y, Gong Z, Shen Y, Yang X, Huang J, Yu F, Li L, Zhu J, Sun L, Fu Y, Kong W. ANK Deficiency-Mediated Cytosolic Citrate Accumulation Promotes Aortic Aneurysm. Circ Res 2024; 135:1175-1192. [PMID: 39513269 DOI: 10.1161/circresaha.124.325152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Disturbed metabolism and transport of citrate play significant roles in various pathologies. However, vascular citrate regulation and its potential role in aortic aneurysm (AA) development remain poorly understood. METHODS Untargeted metabolomics by mass spectrometry was applied to identify upregulated metabolites of the tricarboxylic acid cycle in AA tissues of mice. To investigate the role of citrate and its transporter ANK (progressive ankylosis protein) in AA development, vascular smooth muscle cell (VSMC)-specific Ank-knockout mice were used in both Ang II (angiotensin II)- and CaPO4-induced AA models. RESULTS Citrate was abnormally increased in both human and murine aneurysmal tissues, which was associated with downregulation of ANK, a citrate membrane transporter, in VSMCs. The knockout of Ank in VSMCs promoted AA formation in both Ang II- and CaPO4-induced AA models, while its overexpression inhibited the development of aneurysms. Mechanistically, ANK deficiency in VSMCs caused abnormal cytosolic accumulation of citrate, which was cleaved into acetyl coenzyme A and thus intensified histone acetylation at H3K23, H3K27, and H4K5. Cleavage under target and tagmentation analysis further identified that ANK deficiency-induced histone acetylation activated the transcription of inflammatory genes in VSMCs and thus promoted a citrate-related proinflammatory VSMC phenotype during aneurysm diseases. Accordingly, suppressing citrate cleavage to acetyl coenzyme A downregulated inflammatory gene expression in VSMCs and restricted ANK deficiency-aggravated AA formation. CONCLUSIONS Our studies define the pathogenic role of ANK deficiency-induced cytosolic citrate accumulation in AA pathogenesis and an undescribed citrate-related proinflammatory VSMC phenotype. Targeting ANK-mediated citrate transport may emerge as a novel diagnostic and therapeutic strategy in AA.
Collapse
MESH Headings
- Animals
- Mice
- Citric Acid/metabolism
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/etiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Cytosol/metabolism
- Male
- Cells, Cultured
- Acetylation
- Acetyl Coenzyme A/metabolism
- Disease Models, Animal
- Histones/metabolism
Collapse
Affiliation(s)
- Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, China (L.Y.)
| | - Lin He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, China (H.L., Q.X., J.Z.)
| | - Shiyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qinfeng Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
- Hwamei College of Life and Health Sciences, Zhejiang Wanli University, Ningbo, China (Z.G.)
| | - Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (L.L.)
| | - Junming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Luyang Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling (H.W., Z.L., L.Y., S.Y., Yanjie Li, W.L., Yiran Li, Z.G., Y.S., X.Y., J.H., F.Y., Y.F., W.K.), School of Basic Medical Sciences, Peking University, Beijing, China
| | | |
Collapse
|
9
|
Galey L, Olanrewaju A, Nabi H, Paquette JS, Pouliot F, Audet-Walsh É. PSA, an outdated biomarker for prostate cancer: In search of a more specific biomarker, citrate takes the spotlight. J Steroid Biochem Mol Biol 2024; 243:106588. [PMID: 39025336 DOI: 10.1016/j.jsbmb.2024.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
The prevailing biomarker employed for prostate cancer (PCa) screening and diagnosis is the prostate-specific antigen (PSA). Despite excellent sensitivity, PSA lacks specificity, leading to false positives, unnecessary biopsies and overdiagnosis. Consequently, PSA is increasingly less used by clinicians, thus underscoring the imperative for the identification of new biomarkers. An emerging biomarker in this context is citrate, a molecule secreted by the normal prostate, which has been shown to be inversely correlated with PCa. Here, we discuss about PSA and its usage for PCa diagnosis, its lack of specificity, and the various conditions that can affect its levels. We then provide our vision about what we think would be a valuable addition to our PCa diagnosis toolkit, citrate. We describe the unique citrate metabolic program in the prostate and how this profile is reprogrammed during carcinogenesis. Finally, we summarize the evidence that supports the usage of citrate as a biomarker for PCa diagnosis, as it can be measured in various patient samples and be analyzed by several methods. The unique relationship between citrate and PCa, combined with the stability of citrate levels in other prostate-related conditions and the simplicity of its detection, further accentuates its potential as a biomarker.
Collapse
Affiliation(s)
- Lucas Galey
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Ayokunle Olanrewaju
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hermann Nabi
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada
| | - Jean-Sébastien Paquette
- Laboratoire de recherche et d'innovation en médecine de première ligne (ARIMED), Groupe de médecine de famille universitaire de Saint-Charles-Borromée, CISSS Lanaudière, Saint-Charles-Borromée, QC, Canada; VITAM Research Centre for Sustainable Health, Québec, QC, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Frédéric Pouliot
- Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada; Department of Family Medicine and Emergency Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada; Department of surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Endocrinology - Nephrology Research Axis, Centre de recherche du CHU de Québec - Université Laval, Québec City, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec City, Canada.
| |
Collapse
|
10
|
Xia Y, Zhang MK, Ye JJ, Niu MT, Wang ZY, Dai XY, He ZL, Feng J. Polymeric nanoformulations aimed at cancer metabolism reprogramming with high specificity to inhibit tumor growth. Biomater Sci 2024; 12:5076-5090. [PMID: 39219371 DOI: 10.1039/d4bm00887a] [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
Metabolic disorders of cancer cells create opportunities for metabolic interventions aimed at selectively eliminating cancer cells. Nevertheless, achieving this goal is challenging due to cellular plasticity and metabolic heterogeneity of cancer cells. This study presents a dual-drug-loaded, macrophage membrane-coated polymeric nanovesicle designed to reprogram cancer metabolism with high specificity through integrated extracellular and intracellular interventions. This nanoformulation can target cancer cells and largely reduce their glucose intake, while the fate of intracellular glucose internalized otherwise is redirected at the specially introduced oxidation reaction instead of inherent cancer glycolysis. Meanwhile, it inhibits cellular citrate intake, further reinforcing metabolic intervention. Furthermore, the nanoformulation causes not only H2O2 production, but also NADPH down-regulation, intensifying redox damage to cancer cells. Consequently, this nanoformulation displays highly selective toxicity to cancer cells and minimal harm to normal cells mainly due to metabolic vulnerability of the former. Once administered into tumor-bearing mice, this nanoformulation is found to induce the transformation of pro-tumor tumor associated macrophages into the tumor-suppressive phenotype and completely inhibit tumor growth with favourable biosafety.
Collapse
Affiliation(s)
- Yu Xia
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Ming-Kang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Mei-Ting Niu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Zi-Yang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Xin-Yi Dai
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Zhi-Ling He
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China.
| |
Collapse
|
11
|
Cenigaonandia‐Campillo A, Garcia‐Bautista A, Rio‐Vilariño A, Cebrian A, del Puerto L, Pellicer JA, Gabaldón JA, Pérez‐Sánchez H, Carmena‐Bargueño M, Meroño C, Traba J, Fernandez‐Aceñero MJ, Baños‐Herraiz N, Mozas‐Vivar L, Núñez‐Delicado E, Garcia‐Foncillas J, Aguilera Ó. Vitamin-C-dependent downregulation of the citrate metabolism pathway potentiates pancreatic ductal adenocarcinoma growth arrest. Mol Oncol 2024; 18:2212-2233. [PMID: 38425123 PMCID: PMC11467799 DOI: 10.1002/1878-0261.13616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/17/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
In pancreatic ductal adenocarcinoma (PDAC), metabolic rewiring and resistance to standard therapy are closely associated. PDAC cells show enormous requirements for glucose-derived citrate, the first rate-limiting metabolite in the synthesis of new lipids. Both the expression and activity of citrate synthase (CS) are extraordinarily upregulated in PDAC. However, no previous relationship between gemcitabine response and citrate metabolism has been documented in pancreatic cancer. Here, we report for the first time that pharmacological doses of vitamin C are capable of exerting an inhibitory action on the activity of CS, reducing glucose-derived citrate levels. Moreover, ascorbate targets citrate metabolism towards the de novo lipogenesis pathway, impairing fatty acid synthase (FASN) and ATP citrate lyase (ACLY) expression. Lowered citrate availability was found to be directly associated with diminished proliferation and, remarkably, enhanced gemcitabine response. Moreover, the deregulated citrate-derived lipogenic pathway correlated with a remarkable decrease in extracellular pH through inhibition of lactate dehydrogenase (LDH) and overall reduced glycolytic metabolism. Modulation of citric acid metabolism in highly chemoresistant pancreatic adenocarcinoma, through molecules such as vitamin C, could be considered as a future clinical option to improve patient response to standard chemotherapy regimens.
Collapse
Affiliation(s)
| | - Ana Garcia‐Bautista
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Anxo Rio‐Vilariño
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Arancha Cebrian
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Laura del Puerto
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - José Antonio Pellicer
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - José Antonio Gabaldón
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Horacio Pérez‐Sánchez
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Miguel Carmena‐Bargueño
- Bioinformatics and High‐Performance Computing Research Group (BIO‐HPC), Computer Engineering DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Carolina Meroño
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | - Javier Traba
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones CientíficasUniversidad Autónoma de Madrid (CSIC‐UAM)Spain
- Instituto Universitario de Biología Molecular‐UAM (IUBM‐UAM), Departamento de Biología MolecularUniversidad Autónoma de MadridSpain
| | | | | | - Lorena Mozas‐Vivar
- Preclinical programe START Madrid‐FJD Hospital fundación Jiménez DíazSpain
| | - Estrella Núñez‐Delicado
- Molecular Recognition and Encapsulation Research Group (REM), Health Sciences DepartmentUniversidad Católica de Murcia (UCAM)Spain
| | - Jesús Garcia‐Foncillas
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
| | - Óscar Aguilera
- Translational Oncology Division, Oncohealth InstituteIIS‐Fundación Jimenez Diaz‐UAM (Madrid)Spain
- Universidad Católica de Murcia (UCAM)Spain
| |
Collapse
|
12
|
Gao K, Cao W, He Z, Liu L, Guo J, Dong L, Song J, Wu Y, Zhao Y. Network medicine analysis for dissecting the therapeutic mechanism of consensus TCM formulae in treating hepatocellular carcinoma with different TCM syndromes. Front Endocrinol (Lausanne) 2024; 15:1373054. [PMID: 39211446 PMCID: PMC11357915 DOI: 10.3389/fendo.2024.1373054] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide. Traditional Chinese Medicine (TCM) is widely utilized as an adjunct therapy, improving patient survival and quality of life. TCM categorizes HCC into five distinct syndromes, each treated with specific herbal formulae. However, the molecular mechanisms underlying these treatments remain unclear. Methods We employed a network medicine approach to explore the therapeutic mechanisms of TCM in HCC. By constructing a protein-protein interaction (PPI) network, we integrated genes associated with TCM syndromes and their corresponding herbal formulae. This allowed for a quantitative analysis of the topological and functional relationships between TCM syndromes, HCC, and the specific formulae used for treatment. Results Our findings revealed that genes related to the five TCM syndromes were closely associated with HCC-related genes within the PPI network. The gene sets corresponding to the five TCM formulae exhibited significant proximity to HCC and its related syndromes, suggesting the efficacy of TCM syndrome differentiation and treatment. Additionally, through a random walk algorithm applied to a heterogeneous network, we prioritized active herbal ingredients, with results confirmed by literature. Discussion The identification of these key compounds underscores the potential of network medicine to unravel the complex pharmacological actions of TCM. This study provides a molecular basis for TCM's therapeutic strategies in HCC and highlights specific herbal ingredients as potential leads for drug development and precision medicine.
Collapse
Affiliation(s)
- Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - WanChen Cao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - ZiHao He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - JinCheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Jini Song
- New York Institute of Technology College of Osteopathic Medicine, Arkansas State University, Jonesboro, AR, United States
| | - Yang Wu
- The Research Center for Ubiquitous Computing Systems (CUbiCS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
- The Research Center for Ubiquitous Computing Systems (CUbiCS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Schwertner B, Dahdal G, Jagla W, Grossmann L, Drexler K, Krahn MP, Evert K, Berneburg M, Haferkamp S, Ziegler C, Parkinson EK, Zahn G, Mycielska ME, Gaumann A. Expression of the plasma membrane citrate carrier (pmCiC) in human cancerous tissues-correlation with tumour aggressiveness. Front Cell Dev Biol 2024; 12:1308135. [PMID: 39022761 PMCID: PMC11251970 DOI: 10.3389/fcell.2024.1308135] [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: 10/05/2023] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
We have recently shown that cancer cells of various origins take up extracellular citrate through the plasma membrane citrate carrier (pmCiC), a specific plasma membrane citrate transporter. Extracellular citrate is required to support cancer cell metabolism, in particular fatty acid synthesis, mitochondrial activity, protein synthesis and histone acetylation. In addition, cancer cells tend to acquire a metastatic phenotype in the presence of extracellular citrate. Our recent study also showed that cancer-associated stromal cells synthesise and release citrate and that this process is controlled by cancer cells. In the present study, we evaluated the expression of pmCiC, fibroblast activation protein-α (FAP) and the angiogenesis marker cluster of differentiation 31 (CD31) in human cancer tissues of different origins. In the cohort studied, we found no correlation between disease stage and the expression of FAP or CD31. However, we have identified a clear correlation between pmCiC expression in cancer cells and cancer-associated stroma with tumour stage. It can be concluded that pmCiC is increased in cancer cells and in cancer-supporting cells in the tumour microenvironment at the later stages of cancer development, particularly at the metastatic sites. Therefore, pmCiC expression has the potential to serve as a prognostic marker, although further studies are needed.
Collapse
Affiliation(s)
- Barbara Schwertner
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - George Dahdal
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Jagla
- Institute of Pathology Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Luis Grossmann
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Michael P. Krahn
- Medical Cell Biology, Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Christine Ziegler
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Eric K. Parkinson
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Maria E. Mycielska
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Andreas Gaumann
- Institute of Pathology Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| |
Collapse
|
14
|
Wu J, Liu N, Chen J, Tao Q, Li Q, Li J, Chen X, Peng C. The Tricarboxylic Acid Cycle Metabolites for Cancer: Friend or Enemy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0351. [PMID: 38867720 PMCID: PMC11168306 DOI: 10.34133/research.0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/18/2024] [Indexed: 06/14/2024]
Abstract
The tricarboxylic acid (TCA) cycle is capable of providing sufficient energy for the physiological activities under aerobic conditions. Although tumor metabolic reprogramming places aerobic glycolysis in a dominant position, the TCA cycle remains indispensable for tumor cells as a hub for the metabolic linkage and interconversion of glucose, lipids, and certain amino acids. TCA intermediates such as citrate, α-ketoglutarate, succinate, and fumarate are altered in tumors, and they regulate the tumor metabolism, signal transduction, and immune environment to affect tumorigenesis and tumor progression. This article provides a comprehensive review of the modifications occurring in tumor cells in relation to the intermediates of the TCA cycle, which affects tumor pathogenesis and current therapeutic strategy for therapy through targeting TCA cycle in cancer cells.
Collapse
Affiliation(s)
- Jie Wu
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Nian Liu
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jing Chen
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Qian Tao
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Qiuqiu Li
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jie Li
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Cong Peng
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Vishwa R, BharathwajChetty B, Girisa S, Aswani BS, Alqahtani MS, Abbas M, Hegde M, Kunnumakkara AB. Lipid metabolism and its implications in tumor cell plasticity and drug resistance: what we learned thus far? Cancer Metastasis Rev 2024; 43:293-319. [PMID: 38438800 DOI: 10.1007/s10555-024-10170-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/19/2024] [Indexed: 03/06/2024]
Abstract
Metabolic reprogramming, a hallmark of cancer, allows cancer cells to adapt to their specific energy needs. The Warburg effect benefits cancer cells in both hypoxic and normoxic conditions and is a well-studied reprogramming of metabolism in cancer. Interestingly, the alteration of other metabolic pathways, especially lipid metabolism has also grabbed the attention of scientists worldwide. Lipids, primarily consisting of fatty acids, phospholipids and cholesterol, play essential roles as structural component of cell membrane, signalling molecule and energy reserves. This reprogramming primarily involves aberrations in the uptake, synthesis and breakdown of lipids, thereby contributing to the survival, proliferation, invasion, migration and metastasis of cancer cells. The development of resistance to the existing treatment modalities poses a major challenge in the field of cancer therapy. Also, the plasticity of tumor cells was reported to be a contributing factor for the development of resistance. A number of studies implicated that dysregulated lipid metabolism contributes to tumor cell plasticity and associated drug resistance. Therefore, it is important to understand the intricate reprogramming of lipid metabolism in cancer cells. In this review, we mainly focused on the implication of disturbed lipid metabolic events on inducing tumor cell plasticity-mediated drug resistance. In addition, we also discussed the concept of lipid peroxidation and its crucial role in phenotypic switching and resistance to ferroptosis in cancer cells. Elucidating the relationship between lipid metabolism, tumor cell plasticity and emergence of resistance will open new opportunities to develop innovative strategies and combinatorial approaches for the treatment of cancer.
Collapse
Affiliation(s)
- Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
16
|
Evin D, Evinová A, Baranovičová E, Šarlinová M, Jurečeková J, Kaplán P, Poláček H, Halašová E, Dušenka R, Briš L, Brožová MK, Sivoňová MK. Integrative Metabolomic Analysis of Serum and Selected Serum Exosomal microRNA in Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2024; 25:2630. [PMID: 38473877 DOI: 10.3390/ijms25052630] [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: 01/17/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a lethal disease due to the absence of effective therapies. A more comprehensive understanding of molecular events, encompassing the dysregulation of microRNAs (miRs) and metabolic reprogramming, holds the potential to unveil precise mechanisms underlying mCRPC. This study aims to assess the expression of selected serum exosomal miRs (miR-15a, miR-16, miR-19a-3p, miR-21, and miR-141a-3p) alongside serum metabolomic profiling and their correlation in patients with mCRPC and benign prostate hyperplasia (BPH). Blood serum samples from mCRPC patients (n = 51) and BPH patients (n = 48) underwent metabolome analysis through 1H-NMR spectroscopy. The expression levels of serum exosomal miRs in mCRPC and BPH patients were evaluated using a quantitative real-time polymerase chain reaction (qRT-PCR). The 1H-NMR metabolomics analysis revealed significant alterations in lactate, acetate, citrate, 3-hydroxybutyrate, and branched-chain amino acids (BCAAs, including valine, leucine, and isoleucine) in mCRPC patients compared to BPH patients. MiR-15a, miR-16, miR-19a-3p, and miR-21 exhibited a downregulation of more than twofold in the mCRPC group. Significant correlations were predominantly observed between lactate, citrate, acetate, and miR-15a, miR-16, miR-19a-3p, and miR-21. The importance of integrating metabolome analysis of serum with selected serum exosomal miRs in mCRPC patients has been confirmed, suggesting their potential utility for distinguishing of mCRPC from BPH.
Collapse
Affiliation(s)
- Daniel Evin
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
- Clinic of Nuclear Medicine, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Andrea Evinová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Eva Baranovičová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Miroslava Šarlinová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Jana Jurečeková
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Peter Kaplán
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Hubert Poláček
- Clinic of Nuclear Medicine, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Erika Halašová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Róbert Dušenka
- Clinic of Urology, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lukáš Briš
- Clinic of Urology, Jessenius Faculty of Medicine in Martin, University Hospital in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Martina Knoško Brožová
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Monika Kmeťová Sivoňová
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
17
|
Icard P, Simula L, Zahn G, Alifano M, Mycielska ME. The dual role of citrate in cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188987. [PMID: 37717858 DOI: 10.1016/j.bbcan.2023.188987] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
Citrate is a key metabolite of the Krebs cycle that can also be exported in the cytosol, where it performs several functions. In normal cells, citrate sustains protein acetylation, lipid synthesis, gluconeogenesis, insulin secretion, bone tissues formation, spermatozoid mobility, and immune response. Dysregulation of citrate metabolism is implicated in several pathologies, including cancer. Here we discuss how cancer cells use citrate to sustain their proliferation, survival, and metastatic progression. Also, we propose two paradoxically opposite strategies to reduce tumour growth by targeting citrate metabolism in preclinical models. In the first strategy, we propose to administer in the tumor microenvironment a high amount of citrate, which can then act as a glycolysis inhibitor and apoptosis inducer, whereas the other strategy targets citrate transporters to starve cancer cells from citrate. These strategies, effective in several preclinical in vitro and in vivo cancer models, could be exploited in clinics, particularly to increase sensibility to current anti-cancer agents.
Collapse
Affiliation(s)
- Philippe Icard
- Normandie Univ, UNICAEN, INSERM U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Caen, France; Service of Thoracic Surgery, Cochin Hospital, AP-, HP, 75014, Paris, France.
| | - Luca Simula
- Cochin Institute, INSERM U1016, CNRS UMR8104, University of Paris-Cité, Paris 75014, France
| | | | - Marco Alifano
- Service of Thoracic Surgery, Cochin Hospital, AP-, HP, 75014, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Maria E Mycielska
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
18
|
Pedrosa L, Foguet C, Oliveres H, Archilla I, de Herreros MG, Rodríguez A, Postigo A, Benítez-Ribas D, Camps J, Cuatrecasas M, Castells A, Prat A, Thomson TM, Maurel J, Cascante M. A novel gene signature unveils three distinct immune-metabolic rewiring patterns conserved across diverse tumor types and associated with outcomes. Front Immunol 2022; 13:926304. [PMID: 36119118 PMCID: PMC9479210 DOI: 10.3389/fimmu.2022.926304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Existing immune signatures and tumor mutational burden have only modest predictive capacity for the efficacy of immune check point inhibitors. In this study, we developed an immune-metabolic signature suitable for personalized ICI therapies. A classifier using an immune-metabolic signature (IMMETCOLS) was developed on a training set of 77 metastatic colorectal cancer (mCRC) samples and validated on 4,200 tumors from the TCGA database belonging to 11 types. Here, we reveal that the IMMETCOLS signature classifies tumors into three distinct immune-metabolic clusters. Cluster 1 displays markers of enhanced glycolisis, hexosamine byosinthesis and epithelial-to-mesenchymal transition. On multivariate analysis, cluster 1 tumors were enriched in pro-immune signature but not in immunophenoscore and were associated with the poorest median survival. Its predicted tumor metabolic features suggest an acidic-lactate-rich tumor microenvironment (TME) geared to an immunosuppressive setting, enriched in fibroblasts. Cluster 2 displays features of gluconeogenesis ability, which is needed for glucose-independent survival and preferential use of alternative carbon sources, including glutamine and lipid uptake/β-oxidation. Its metabolic features suggest a hypoxic and hypoglycemic TME, associated with poor tumor-associated antigen presentation. Finally, cluster 3 is highly glycolytic but also has a solid mitochondrial function, with concomitant upregulation of glutamine and essential amino acid transporters and the pentose phosphate pathway leading to glucose exhaustion in the TME and immunosuppression. Together, these findings suggest that the IMMETCOLS signature provides a classifier of tumors from diverse origins, yielding three clusters with distinct immune-metabolic profiles, representing a new predictive tool for patient selection for specific immune-metabolic therapeutic approaches.
Collapse
Affiliation(s)
- Leire Pedrosa
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Carles Foguet
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Helena Oliveres
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Iván Archilla
- Pathology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Marta García de Herreros
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Adela Rodríguez
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Antonio Postigo
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Group of Transcriptional Regulation of Gene Expression, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institución Catalana de Investigación y Estudios Avanzados (ICREA) and Department of Biomedicine, Universitat de Barcelona, Barcelona, Spain
| | | | - Jordi Camps
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Gastrointestinal Oncology Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Cuatrecasas
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Pathology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Antoni Castells
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Gastrointestinal Oncology Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Aleix Prat
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Timothy M. Thomson
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Cell Biology, Molecular Biology Institute, National Research Council (IBMB-CSIC), Barcelona, Spain
- Universidad Peruana Cayetano Heredia, Lima, Peru
- *Correspondence: Timothy M. Thomson, ; Joan Maurel, ; Marta Cascante,
| | - Joan Maurel
- Medical Oncology Department, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Gastrointestinal Oncology Department, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- *Correspondence: Timothy M. Thomson, ; Joan Maurel, ; Marta Cascante,
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- *Correspondence: Timothy M. Thomson, ; Joan Maurel, ; Marta Cascante,
| |
Collapse
|
19
|
Pion E, Karnosky J, Boscheck S, Wagner BJ, Schmidt KM, Brunner SM, Schlitt HJ, Aung T, Hackl C, Haerteis S. 3D In Vivo Models for Translational Research on Pancreatic Cancer: The Chorioallantoic Membrane (CAM) Model. Cancers (Basel) 2022; 14:cancers14153733. [PMID: 35954398 PMCID: PMC9367548 DOI: 10.3390/cancers14153733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The 5-year overall survival rate for all stages of pancreatic cancer is relatively low at about only 6%. As a result of this exceedingly poor prognosis, new research models are necessary to investigate this highly malignant cancer. One model that has been used extensively for a vast variety of different cancers is the chorioallantoic membrane (CAM) model. It is based on an exceptionally vascularized membrane that develops within fertilized chicken eggs and can be used for the grafting and analysis of tumor tissue. The aim of the study was to summarize already existing works on pancreatic ductal adenocarcinoma (PDAC) and the CAM model. The results were subdivided into different categories that include drug testing, angiogenesis, personalized medicine, modifications of the model, and further developments to help improve the unfavorable prognosis of this disease. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with adverse outcomes that have barely improved over the last decade. About half of all patients present with metastasis at the time of diagnosis, and the 5-year overall survival rate across all stages is only 6%. Innovative in vivo research models are necessary to combat this cancer and to discover novel treatment strategies. The chorioallantoic membrane (CAM) model represents one 3D in vivo methodology that has been used in a large number of studies on different cancer types for over a century. This model is based on a membrane formed within fertilized chicken eggs that contain a dense network of blood vessels. Because of its high cost-efficiency, simplicity, and versatility, the CAM model appears to be a highly valuable research tool in the pursuit of gaining more in-depth insights into PDAC. A summary of the current literature on the usage of the CAM model for the investigation of PDAC was conducted and subdivided into angiogenesis, drug testing, modifications, personalized medicine, and further developments. On this comprehensive basis, further research should be conducted on PDAC in order to improve the abysmal prognosis of this malignant disease.
Collapse
Affiliation(s)
- Eric Pion
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
| | - Julia Karnosky
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Sofie Boscheck
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
| | - Benedikt J. Wagner
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Katharina M. Schmidt
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Stefan M. Brunner
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Hans J. Schlitt
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, 94469 Deggendorf, Germany
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
- Correspondence:
| |
Collapse
|
20
|
Han Y, Yoo HJ, Jee SH, Lee JH. High serum levels of L-carnitine and citric acid negatively correlated with alkaline phosphatase are detectable in Koreans before gastric cancer onset. Metabolomics 2022; 18:62. [PMID: 35900644 DOI: 10.1007/s11306-022-01922-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/14/2022] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Monitoring metabolic biomarkers could be utilized as an effective tool for the early detection of gastric cancer (GC) risk. OBJECTIVE We aimed to discover predictive serum biomarkers for GC and investigate biomarker-related metabolism. METHODS Subjects were randomly selected from the Korean Cancer Prevention Study-II cohort and matched by age and sex. We analyzed baseline serum samples of 160 subjects (discovery set; control and GC occurrence group, 80 each) via nontargeted screening. Identified putative biomarkers were validated in baseline serum samples of 140 subjects (validation set; control and GC occurrence group, 70 each) using targeted metabolites analysis. RESULTS The final analysis was conducted on the discovery set (control, n = 52 vs. GC occurrence, n = 50) and the validation set (control, n = 43 vs. GC occurrence, n = 44) applying exclusion conditions. Eighteen putative metabolite sets differed between two groups found on nontargeted metabolic screening. We focused on fatty acid-related energy metabolism. In targeted analysis, levels of decanoyl-L-carnitine (p = 0.019), L-carnitine (p = 0.033), and citric acid (p = 0.025) were significantly lower in the GC occurrence group, even after adjusting for age, sex, and smoking status. Additionally, L-carnitine and citric acid were confirmed to have an independently significant relationship to GC development. Notably, alkaline phosphatase showed a significant correlation with these two biomarkers. CONCLUSION Changes in serum L-carnitine and citric acid levels that may result from alterations of fatty-acid-related energy metabolism are expected to be valuable biomarkers for the early diagnosis of GC risk.
Collapse
Affiliation(s)
- Youngmin Han
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye Jin Yoo
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Jong Ho Lee
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
21
|
Deep Learning-Based Image Analysis for the Quantification of Tumor-Induced Angiogenesis in the 3D In Vivo Tumor Model—Establishment and Addition to Laser Speckle Contrast Imaging (LSCI). Cells 2022; 11:cells11152321. [PMID: 35954165 PMCID: PMC9367525 DOI: 10.3390/cells11152321] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/17/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: angiogenesis plays an important role in the growth and metastasis of tumors. We established the CAM assay application, an image analysis software of the IKOSA platform by KML Vision, for the quantification of blood vessels with the in ovo chorioallantoic membrane (CAM) model. We added this proprietary deep learning algorithm to the already established laser speckle contrast imaging (LSCI). (2) Methods: angiosarcoma cell line tumors were grafted onto the CAM. Angiogenesis was measured at the beginning and at the end of tumor growth with both measurement methods. The CAM assay application was trained to enable the recognition of in ovo CAM vessels. Histological stains of the tissue were performed and gluconate, an anti-angiogenic substance, was applied to the tumors. (3) Results: the angiosarcoma cells formed tumors on the CAM that appeared to stay vital and proliferated. An increase in perfusion was observed using both methods. The CAM assay application was successfully established in the in ovo CAM model and anti-angiogenic effects of gluconate were observed. (4) Conclusions: the CAM assay application appears to be a useful method for the quantification of angiogenesis in the CAM model and gluconate could be a potential treatment of angiosarcomas. Both aspects should be evaluated in further research.
Collapse
|
22
|
Drexler K, Schwertner B, Haerteis S, Aung T, Berneburg M, Geissler EK, Mycielska ME, Haferkamp S. The Role of Citrate Homeostasis in Merkel Cell Carcinoma Pathogenesis. Cancers (Basel) 2022; 14:cancers14143425. [PMID: 35884486 PMCID: PMC9325124 DOI: 10.3390/cancers14143425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Merkel cell carcinoma (MCC) is a rare but highly aggressive skin cancer. Despite important progress, overall understanding of the events that drive MCC carcinogenesis remains incomplete. We discovered that the plasma membrane citrate transporter (pmCiC) is upregulated in Merkel cell carcinoma cell lines. Cancer cells import extracellular citrate via pmCiC to support their metabolism, which is critical to support proliferation and metastatic spread. In this study, we show that inhibition of pmCiC can decrease the growth rate of Merkel cell carcinoma cell lines. Targeting pmCiC and thereby the tumor metabolism should be considered further as a potential anti-cancer therapy. Abstract Merkel cell carcinoma (MCC) is a rare but highly aggressive tumor of the skin with a poor prognosis. The factors driving this cancer must be better understood in order to discover novel targets for more effective therapies. In the search for targets, we followed our interest in citrate as a central and critical metabolite linked to fatty acid synthesis in cancer development. A key to citrate uptake in cancer cells is the high expression of the plasma membrane citrate transporter (pmCiC), which is upregulated in the different adenocarcinoma types tested so far. In this study, we show that the pmCiC is also highly expressed in Merkel cell carcinoma cell lines by western blot and human tissues by immunohistochemistry staining. In the presence of extracellular citrate, MCC cells show an increased proliferation rate in vitro; a specific pmCiC inhibitor (Na+-gluconate) blocks this citrate-induced proliferation. Furthermore, the 3D in vivo Chick Chorioallantoic Membrane (CAM) model showed that the application of Na+-gluconate also decreases Merkel cell carcinoma growth. Based on our results, we conclude that pmCiC and extracellular citrate uptake should be considered further as a potential novel target for the treatment of Merkel cell carcinoma.
Collapse
Affiliation(s)
- Konstantin Drexler
- Department of Dermatology, University Medical Center, 93053 Regensburg, Germany; (B.S.); (M.B.); (S.H.)
- Correspondence: ; Tel.: +49-941-944-9603; Fax: +49-941-944-9525
| | - Barbara Schwertner
- Department of Dermatology, University Medical Center, 93053 Regensburg, Germany; (B.S.); (M.B.); (S.H.)
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (S.H.); (T.A.)
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (S.H.); (T.A.)
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, 94469 Deggendorf, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Center, 93053 Regensburg, Germany; (B.S.); (M.B.); (S.H.)
| | - Edward K. Geissler
- Department of Surgery, Section of Experimental Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Maria E. Mycielska
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany;
| | - Sebastian Haferkamp
- Department of Dermatology, University Medical Center, 93053 Regensburg, Germany; (B.S.); (M.B.); (S.H.)
| |
Collapse
|
23
|
Kim SY, Kim D, Kim J, Ko HY, Kim WJ, Park Y, Lee HW, Han DH, Kim KS, Park S, Lee M, Yun M. Extracellular Citrate Treatment Induces HIF1α Degradation and Inhibits the Growth of Low-Glycolytic Hepatocellular Carcinoma under Hypoxia. Cancers (Basel) 2022; 14:3355. [PMID: 35884416 PMCID: PMC9315704 DOI: 10.3390/cancers14143355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 12/04/2022] Open
Abstract
HCC is well known for low glycolysis in the tumors, whereas hypoxia induces glycolytic phenotype and tumor progression. This study was conducted to evaluate the expression of SLCs in human HCCs and investigated whether extracellular nutrient administration related to SLCs in low-glycolytic HCC can prevent hypoxic tumor progression. SLCs expression was screened according to the level of glycolysis in HCCs. Then, whether extracellular nutrient treatment can affect hypoxic tumor progression, as well as the mechanisms, were evaluated in an in vitro cell line and an in vivo animal model. Low-glycolytic HCCs showed high SLC13A5/NaCT and SLC16A1/MCT1 but low SLC2A1/GLUT1 and HIF1α/HIF1α expression. Especially, high SLC13A5 expression was significantly associated with good overall survival in the Cancer Genome Atlas (TCGA) database. In HepG2 cells with the highest NaCT expression, extracellular citrate treatment upon hypoxia induced HIF1α degradation, which led to reduced glycolysis and cellular proliferation. Finally, in HepG2-animal models, the citrate-treated group showed smaller tumor with less hypoxic areas than the vehicle-treated group. In patients with HCC, SLC13A5/NaCT is an important SLC, which is associated with low glycolysis and good prognosis. Extracellular citrate treatment induced the failure of metabolic adaptation to hypoxia and tumor growth inhibition, which can be a potential therapeutic strategy in HCCs.
Collapse
Affiliation(s)
- Seon Yoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| | - Dongwoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| | - Jisu Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| | - Hae Young Ko
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| | - Won Jin Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea;
| | - Youngjoo Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| | - Hye Won Lee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Dai Hoon Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (D.H.H.); (K.S.K.)
| | - Kyung Sik Kim
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (D.H.H.); (K.S.K.)
| | - Sunghyouk Park
- Department of Manufacturing Pharmacy, Natural Product Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Korea;
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea;
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.K.); (D.K.); (J.K.); (H.Y.K.); (Y.P.)
| |
Collapse
|
24
|
Cancer Cell Metabolism. Int J Mol Sci 2022; 23:ijms23137210. [PMID: 35806215 PMCID: PMC9266817 DOI: 10.3390/ijms23137210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 12/10/2022] Open
|
25
|
Ganapathy V, Haferkamp S, Parkinson EK, Mycielska ME. Editorial: Metabolite and Nutrient Transporters in Cancer-Cell Metabolism: Role in Cancer Progression and Metastasis. Front Cell Dev Biol 2022; 10:885717. [PMID: 35547821 PMCID: PMC9081672 DOI: 10.3389/fcell.2022.885717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/07/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- *Correspondence: Vadivel Ganapathy, ; Sebastian Haferkamp, ; Eric K. Parkinson, ; Maria E. Mycielska,
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
- *Correspondence: Vadivel Ganapathy, ; Sebastian Haferkamp, ; Eric K. Parkinson, ; Maria E. Mycielska,
| | - Eric K. Parkinson
- Centre for Immunobiology and Regenerative Medicine, Queen Mary University of London, London, United Kingdom
- *Correspondence: Vadivel Ganapathy, ; Sebastian Haferkamp, ; Eric K. Parkinson, ; Maria E. Mycielska,
| | - Maria E. Mycielska
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
- *Correspondence: Vadivel Ganapathy, ; Sebastian Haferkamp, ; Eric K. Parkinson, ; Maria E. Mycielska,
| |
Collapse
|
26
|
Parkinson EK, Prime SS. Oral Senescence: From Molecular Biology to Clinical Research. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.822397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cellular senescence is an irreversible cell cycle arrest occurring following multiple rounds of cell division (replicative senescence) or in response to cellular stresses such as ionizing radiation, signaling imbalances and oxidative damage (stress-induced premature senescence). Even very small numbers of senescent cells can be deleterious and there is evidence that senescent cells are instrumental in a number of oral pathologies including cancer, oral sub mucous fibrosis and the side effects of cancer therapy. In addition, senescent cells are present and possibly important in periodontal disease and other chronic inflammatory conditions of the oral cavity. However, senescence is a double-edged sword because although it operates as a suppressor of malignancy in pre-malignant epithelia, senescent cells in the neoplastic environment promote tumor growth and progression. Many of the effects of senescent cells are dependent on the secretion of an array of diverse therapeutically targetable proteins known as the senescence-associated secretory phenotype. However, as senescence may have beneficial roles in wound repair, preventing fibrosis and stem cell activation the clinical exploitation of senescent cells is not straightforward. Here, we discuss biological mechanisms of senescence and we review the current approaches to target senescent cells therapeutically, including senostatics and senolytics which are entering clinical trials.
Collapse
|
27
|
Citrate transporter inhibitors: possible new anticancer agents. Future Med Chem 2022; 14:665-679. [PMID: 35357238 DOI: 10.4155/fmc-2021-0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The culmination of 80+ years of cancer research implicates the aberrant metabolism in tumor cells as a root cause of pathogenesis. Citrate is an essential molecule in intermediary metabolism, and its amplified availability to critical pathways in cancer cells via citrate transporters confers a high rate of cancer cell growth and proliferation. Inhibiting the plasma membrane and mitochondrial citrate transporters - whether individually, in combination, or partnered with complementary metabolic targets - in order to combat cancer may prove to be a consequential chemotherapeutic strategy. This review aims to summarize the use of different classes of citrate transporter inhibitors for anticancer activity, either individually or as part of a cocktail.
Collapse
|
28
|
Metabolic Alterations in Cellular Senescence: The Role of Citrate in Ageing and Age-Related Disease. Int J Mol Sci 2022; 23:ijms23073652. [PMID: 35409012 PMCID: PMC8998297 DOI: 10.3390/ijms23073652] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Recent mouse model experiments support an instrumental role for senescent cells in age-related diseases and senescent cells may be causal to certain age-related pathologies. A strongly supported hypothesis is that extranuclear chromatin is recognized by the cyclic GMP–AMP synthase-stimulator of interferon genes pathway, which in turn leads to the induction of several inflammatory cytokines as part of the senescence-associated secretory phenotype. This sterile inflammation increases with chronological age and age-associated disease. More recently, several intracellular and extracellular metabolic changes have been described in senescent cells but it is not clear whether any of them have functional significance. In this review, we highlight the potential effect of dietary and age-related metabolites in the modulation of the senescent phenotype in addition to discussing how experimental conditions may influence senescent cell metabolism, especially that of energy regulation. Finally, as extracellular citrate accumulates following certain types of senescence, we focus on the recently reported role of extracellular citrate in aging and age-related pathologies. We propose that citrate may be an active component of the senescence-associated secretory phenotype and via its intake through the diet may even contribute to the cause of age-related disease.
Collapse
|
29
|
Liu X, Hoft DF, Peng G. Tumor microenvironment metabolites directing T cell differentiation and function. Trends Immunol 2022; 43:132-147. [PMID: 34973923 PMCID: PMC8810659 DOI: 10.1016/j.it.2021.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 02/03/2023]
Abstract
Metabolic reprogramming of cancer cells creates a unique tumor microenvironment (TME) characterized by the limited availability of nutrients, which subsequently affects the metabolism, differentiation, and function of tumor-infiltrating T lymphocytes (TILs). TILs can also be inhibited by tumor-derived metabolic waste products and low oxygen. Therefore, a thorough understanding of how such unique metabolites influence mammalian T cell differentiation and function can inform novel anticancer therapeutic approaches. Here, we highlight the importance of these metabolites in modulating various T cell subsets within the TME, dissecting how these changes might alter clinical outcomes. We explore potential TME metabolic determinants that might constitute candidate targets for cancer immunotherapies, ideally leading to future strategies for reprogramming tumor metabolism to potentiate anticancer T cell functions.
Collapse
Affiliation(s)
- Xia Liu
- Division of Infectious Diseases, Allergy and Immunology and Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Daniel F Hoft
- Division of Infectious Diseases, Allergy and Immunology and Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA; Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, MO 63104, USA
| | - Guangyong Peng
- Division of Infectious Diseases, Allergy and Immunology and Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA; Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, MO 63104, USA.
| |
Collapse
|
30
|
Branco JR, Esteves AM, Imbroisi Filho R, Demaria TM, Lisboa PC, Lopes BP, Moura EG, Zancan P, Sola-Penna M. Citrate enrichment in a Western diet reduces weight gain via browning of adipose tissues without resolving diet-induced insulin resistance in mice. Food Funct 2022; 13:10947-10955. [DOI: 10.1039/d2fo02011d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Citrate, a major component of processed foods, reduces weight gain without resolving insulin resistance.
Collapse
Affiliation(s)
- Jessica Ristow Branco
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Amanda Moreira Esteves
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Ricardo Imbroisi Filho
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Thainá M. Demaria
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Patricia C. Lisboa
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Bruna Pereira Lopes
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Egberto G. Moura
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Patricia Zancan
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Mauro Sola-Penna
- The MetaboliZSm’ GrouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| |
Collapse
|
31
|
Zhao Y, Liu X, Si F, Huang L, Gao A, Lin W, Hoft DF, Shao Q, Peng G. Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2101553. [PMID: 34747157 PMCID: PMC8728847 DOI: 10.1002/advs.202101553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/04/2021] [Indexed: 05/17/2023]
Abstract
Metabolic disorder is one of the hallmarks of cancers, and reprogramming of metabolism is becoming a novel strategy for cancer treatment. Citrate is a key metabolite and critical metabolic regulator linking glycolysis and lipid metabolism in cellular energy homeostasis. Here it is reported that citrate treatment (both sodium citrate and citric acid) significantly suppresses tumor cell proliferation and growth in various tumor types. Mechanistically, citrate promotes excessive lipid biosynthesis and induces disruption of lipid metabolism in tumor cells, resulting in tumor cell senescence and growth inhibition. Furthermore, ATM-associated DNA damage response cooperates with MAPK and mTOR signaling pathways to control citrate-induced tumor cell growth arrest and senescence. In vivo studies further demonstrate that citrate administration dramatically inhibits tumor growth and progression in a colon cancer xenograft model. Importantly, citrate administration combined with the conventional chemotherapy drugs exhibits synergistic antitumor effects in vivo in the colon cancer models. These results clearly indicate that citrate can reprogram lipid metabolism and cell fate in cancer cells, and targeting citrate can be a promising therapeutic strategy for tumor treatment.
Collapse
Affiliation(s)
- Yangjing Zhao
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Xia Liu
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Fusheng Si
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Lan Huang
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Aiqin Gao
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Wenli Lin
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Daniel F. Hoft
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
- Department of Molecular Microbiology & ImmunologySaint Louis University School of MedicineSaint LouisMO63104USA
| | - Qixiang Shao
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
| | - Guangyong Peng
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
- Department of Molecular Microbiology & ImmunologySaint Louis University School of MedicineSaint LouisMO63104USA
| |
Collapse
|
32
|
Parkinson EK, Adamski J, Zahn G, Gaumann A, Flores-Borja F, Ziegler C, Mycielska ME. Extracellular citrate and metabolic adaptations of cancer cells. Cancer Metastasis Rev 2021; 40:1073-1091. [PMID: 34932167 PMCID: PMC8825388 DOI: 10.1007/s10555-021-10007-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022]
Abstract
It is well established that cancer cells acquire energy via the Warburg effect and oxidative phosphorylation. Citrate is considered to play a crucial role in cancer metabolism by virtue of its production in the reverse Krebs cycle from glutamine. Here, we review the evidence that extracellular citrate is one of the key metabolites of the metabolic pathways present in cancer cells. We review the different mechanisms by which pathways involved in keeping redox balance respond to the need of intracellular citrate synthesis under different extracellular metabolic conditions. In this context, we further discuss the hypothesis that extracellular citrate plays a role in switching between oxidative phosphorylation and the Warburg effect while citrate uptake enhances metastatic activities and therapy resistance. We also present the possibility that organs rich in citrate such as the liver, brain and bones might form a perfect niche for the secondary tumour growth and improve survival of colonising cancer cells. Consistently, metabolic support provided by cancer-associated and senescent cells is also discussed. Finally, we highlight evidence on the role of citrate on immune cells and its potential to modulate the biological functions of pro- and anti-tumour immune cells in the tumour microenvironment. Collectively, we review intriguing evidence supporting the potential role of extracellular citrate in the regulation of the overall cancer metabolism and metastatic activity.
Collapse
Affiliation(s)
- E Kenneth Parkinson
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London, E1 2AD, UK.
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Department of Experimental Genetics, Technical University of Munich, Munich, Germany.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Andreas Gaumann
- Institute of Pathology Kaufbeuren-Ravensburg, 87600, Kaufbeuren, Germany
| | - Fabian Flores-Borja
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London, E1 2AD, UK
| | - Christine Ziegler
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstrasse 31, 93053, Regensburg, Germany
| | - Maria E Mycielska
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstrasse 31, 93053, Regensburg, Germany.
| |
Collapse
|
33
|
Juan Z, Qing Z, Yongping L, Qian L, Wu W, Wen Y, Tong J, Ding B. Probiotics for the Treatment of Docetaxel-Related Weight Gain of Breast Cancer Patients-A Single-Center, Randomized, Double-Blind, and Placebo-Controlled Trial. Front Nutr 2021; 8:762929. [PMID: 34926547 PMCID: PMC8675585 DOI: 10.3389/fnut.2021.762929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Docetaxel is an important chemotherapy-agent for breast cancer treatment. One of its side-effects is weight gain, which increases the all-cause mortality rate. Considering gut microbiota is one important factor for weight regulation, we hypothesized that probiotics could be potentially used to reduce the docetaxel-related weight gain in breast cancer patients. Methods: From 10/8/2018 to 10/17/2019, 100 breast cancer (Stage I-III) patients underwent four cycles of docetaxel-based chemotherapy were enrolled and randomly assigned to receive probiotics (Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis) or placebo (supplementary material of the probiotics capsule) treatment for 84 days with three capsules per time, twice/day. The primary outcome: the changes in body weight and body-fat percentage of the patients were measured by a designated physician using a fat analyzer, and the secondary outcomes: the fasting insulin, plasma glucose, and lipids were directly obtained from the Hospital Information System (HIS); The metabolites were measured using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS); The fecal microbiome was analyzed using bacterial 16S ribosomal RNA (rRNA) gene sequence. All indicators were measured 1 day before the first cycle of docetaxel-based chemotherapy and 21 days after the last cycle of docetaxel-based chemotherapy. Results: Compared with the placebo group, the probiotic group showed significantly smaller changes in body weight (Mean [SD] 0.77 [2.58] vs. 2.70 [3.08], P = 0.03), body-fat percentage (Mean [SD] 0.04 [1.14] vs. 3.86 [11.09], P = 0.02), and low density lipoprotein (LDL) (Mean [SD]-0.05[0.68] vs. 0.39 [0.58], P = 0.002). Moreover, five of the 340 detected plasma metabolites showed significant differences between the two groups. The change of biliverdin dihydrochloride (B = -0.724, P = 0.02) was inverse correlated with weight gain. One strain of the phylum and three strains of the genus were detected to be significantly different between the two groups. Also, the changes of Bacteroides (B = -0.917, P < 0.001) and Anaerostipes (B = -0.894, P < 0.001) were inverse correlated with the change of LDL. Conclusions: Probiotics supplement during docetaxel-based chemotherapy for breast cancer treatment may help to reduce the increase in body weight, body-fat percentage, plasma LDL, and minimize the metabolic changes and gut dysbacteriosis. Clinical Trial Registration: http://www.chictr.org.cn/showproj.aspx?proj=24294, ChiCTR-INQ-17014181.
Collapse
Affiliation(s)
- Zhang Juan
- Department of Breast Surgery, Tangshan People's Hospital, Tangshan, Hebei, China
| | - Zhang Qing
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liang Yongping
- Department of Medical Imaging (Ultrasound), Tangshan Central Hospital, Tangshan, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yanguang Wen
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jianbin Tong
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha, China
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Boni Ding
- Department of Breast and Thyroid Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Kumar A, Cordes T, Thalacker-Mercer AE, Pajor AM, Murphy AN, Metallo CM. NaCT/SLC13A5 facilitates citrate import and metabolism under nutrient-limited conditions. Cell Rep 2021; 36:109701. [PMID: 34525352 PMCID: PMC8500708 DOI: 10.1016/j.celrep.2021.109701] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/19/2021] [Accepted: 08/20/2021] [Indexed: 01/11/2023] Open
Abstract
Citrate lies at a critical node of metabolism, linking tricarboxylic acid metabolism and lipogenesis via acetyl-coenzyme A. Recent studies have observed that deficiency of the sodium-dependent citrate transporter (NaCT), encoded by SLC13A5, dysregulates hepatic metabolism and drives pediatric epilepsy. To examine how NaCT contributes to citrate metabolism in cells relevant to the pathophysiology of these diseases, we apply 13C isotope tracing to SLC13A5-deficient hepatocellular carcinoma (HCC) cells and primary rat cortical neurons. Exogenous citrate appreciably contributes to intermediary metabolism only under hypoxic conditions. In the absence of glutamine, citrate supplementation increases de novo lipogenesis and growth of HCC cells. Knockout of SLC13A5 in Huh7 cells compromises citrate uptake and catabolism. Citrate supplementation rescues Huh7 cell viability in response to glutamine deprivation or Zn2+ treatment, and NaCT deficiency mitigates these effects. Collectively, these findings demonstrate that NaCT-mediated citrate uptake is metabolically important under nutrient-limited conditions and may facilitate resistance to metal toxicity.
Collapse
Affiliation(s)
- Avi Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thekla Cordes
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna E Thalacker-Mercer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ana M Pajor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anne N Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
35
|
Karen-Ng LP, James EL, Stephen A, Bennett MH, Mycielska ME, Parkinson EK. The Extracellular Metabolome Stratifies Low and High Risk Potentially Premalignant Oral Keratinocytes and Identifies Citrate as a Potential Non-Invasive Marker of Tumour Progression. Cancers (Basel) 2021; 13:cancers13164212. [PMID: 34439366 PMCID: PMC8394991 DOI: 10.3390/cancers13164212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The early detection of oral cancer is a high priority, as improvements in this area could lead to greater cure rates and reduced disability due to extensive surgery. Oral cancer is very difficult to detect in over 70% of cases as it develops unseen until quite advanced, sometimes rapidly. Therefore, the development of markers in body fluids (liquid biopsies) indicative of cancerous changes have a high priority. We show here that small molecules called metabolites can distinguish between non-diseased oral cells and two types of cells found in oral cells on the road to cancer. Although our investigation is preliminary, some of the metabolites have already been detected in the saliva (split) of oral cancer patients, and could eventually help detect oral cancer development at an earlier stage. Abstract Premalignant oral lesions (PPOLs) which bypass senescence (IPPOL) have a much greater probability of progressing to malignancy, but pre-cancerous fields also contain mortal PPOL keratinocytes (MPPOL) that possess tumour-promoting properties. To identify metabolites that could potentially separate IPPOL, MPPOL and normal oral keratinocytes non-invasively in vivo, we conducted an unbiased screen of their conditioned medium. MPPOL keratinocytes showed elevated levels of branch-chain amino acid, lipid, prostaglandin, and glutathione metabolites, some of which could potentially be converted into volatile compounds by oral bacteria and detected in breath analysis. Extracellular metabolites were generally depleted in IPPOL, and only six were elevated, but some metabolites distinguishing IPPOL from MPPOL have been associated with progression to oral squamous cell carcinoma (OSCC) in vivo. One of the metabolites elevated in IPPOL relative to the other groups, citrate, was confirmed by targeted metabolomics and, interestingly, has been implicated in cancer growth and metastasis. Although our investigation is preliminary, some of the metabolites described here are detectable in the saliva of oral cancer patients, albeit at a more advanced stage, and could eventually help detect oral cancer development earlier.
Collapse
Affiliation(s)
- Lee Peng Karen-Ng
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Emma Louise James
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
| | - Abish Stephen
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
| | - Mark Henry Bennett
- Department of Life Science, South Kensington Campus, Imperial College London, London SW7 2AZ, UK;
| | - Maria Elzbieta Mycielska
- Department of Surgery, University Medical Center, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany;
| | - Eric Kenneth Parkinson
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
- Correspondence: ; Tel.: +44-(0)207-882-7185 or +44-(0)78546536
| |
Collapse
|
36
|
Branco JR, Esteves AM, Leandro JGB, Demaria TM, Godoi V, Marette A, Valença HDM, Lanzetti M, Peyot ML, Farfari S, Prentki M, Zancan P, Sola-Penna M. Dietary citrate acutely induces insulin resistance and markers of liver inflammation in mice. J Nutr Biochem 2021; 98:108834. [PMID: 34371126 DOI: 10.1016/j.jnutbio.2021.108834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Citrate is widely used as a food additive being part of virtually all processed foods. Although considered inert by most of the regulatory agencies in the world, plasma citrate has been proposed to play immunometabolic functions in multiple tissues through altering a plethora of cellular pathways. Here, we used a short-term alimentary intervention (24 hours) with standard chow supplemented with citrate in amount corresponding to that found in processed foods to evaluate its effects on glucose homeostasis and liver physiology in C57BL/6J mice. Animals supplemented with dietary citrate showed glucose intolerance and insulin resistance as revealed by glucose and insulin tolerance tests. Moreover, animals supplemented with citrate in their food displayed fed and fasted hyperinsulinemia and enhanced insulin secretion during an oral glucose tolerance test. Citrate treatment also amplified glucose-induced insulin secretion in vitro in INS1-E cells. Citrate supplemented animals had increased liver PKCα activity and altered phosphorylation at serine or threonine residues of components of insulin signaling including IRS-1, Akt, GSK-3 and FoxO1. Furthermore, citrate supplementation enhanced the hepatic expression of lipogenic genes suggesting increased de novo lipogenesis, a finding that was reproduced after citrate treatment of hepatic FAO cells. Finally, liver inflammation markers were higher in citrate supplemented animals. Overall, the results demonstrate that dietary citrate supplementation in mice causes hyperinsulinemia and insulin resistance both in vivo and in vitro, and therefore call for a note of caution on the use of citrate as a food additive given its potential role in metabolic dysregulation.
Collapse
Affiliation(s)
- Jessica Ristow Branco
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Amanda Moreira Esteves
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - João Gabriel Bernardo Leandro
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM) Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thainá M Demaria
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM) Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Vilma Godoi
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratório de Enzimologia e Controle do Metabolismo (LabECoM) Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Departamento de Ciências Morfológicas, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - André Marette
- Department of Medicine, Quebec Heart and Lung Institute, Hôpital Laval, Pavillon Marguerite d'Youville, Québec, Canada
| | - Helber da Maia Valença
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manuella Lanzetti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marie-Line Peyot
- Molecular Nutrition Unit, Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), department of Nutrition, Université de Montréal, Montréal, Canada
| | - Salah Farfari
- Molecular Nutrition Unit, Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), department of Nutrition, Université de Montréal, Montréal, Canada
| | - Marc Prentki
- Molecular Nutrition Unit, Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), department of Nutrition, Université de Montréal, Montréal, Canada
| | - Patricia Zancan
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM) Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
37
|
Understanding the Central Role of Citrate in the Metabolism of Cancer Cells and Tumors: An Update. Int J Mol Sci 2021; 22:ijms22126587. [PMID: 34205414 PMCID: PMC8235534 DOI: 10.3390/ijms22126587] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Citrate plays a central role in cancer cells’ metabolism and regulation. Derived from mitochondrial synthesis and/or carboxylation of α-ketoglutarate, it is cleaved by ATP-citrate lyase into acetyl-CoA and oxaloacetate. The rapid turnover of these molecules in proliferative cancer cells maintains a low-level of citrate, precluding its retro-inhibition on glycolytic enzymes. In cancer cells relying on glycolysis, this regulation helps sustain the Warburg effect. In those relying on an oxidative metabolism, fatty acid β-oxidation sustains a high production of citrate, which is still rapidly converted into acetyl-CoA and oxaloacetate, this latter molecule sustaining nucleotide synthesis and gluconeogenesis. Therefore, citrate levels are rarely high in cancer cells. Resistance of cancer cells to targeted therapies, such as tyrosine kinase inhibitors (TKIs), is frequently sustained by aerobic glycolysis and its key oncogenic drivers, such as Ras and its downstream effectors MAPK/ERK and PI3K/Akt. Remarkably, in preclinical cancer models, the administration of high doses of citrate showed various anti-cancer effects, such as the inhibition of glycolysis, the promotion of cytotoxic drugs sensibility and apoptosis, the neutralization of extracellular acidity, and the inhibition of tumors growth and of key signalling pathways (in particular, the IGF-1R/AKT pathway). Therefore, these preclinical results support the testing of the citrate strategy in clinical trials to counteract key oncogenic drivers sustaining cancer development and resistance to anti-cancer therapies.
Collapse
|
38
|
Drexler K, Schmidt KM, Jordan K, Federlin M, Milenkovic VM, Liebisch G, Artati A, Schmidl C, Madej G, Tokarz J, Cecil A, Jagla W, Haerteis S, Aung T, Wagner C, Kolodziejczyk M, Heinke S, Stanton EH, Schwertner B, Riegel D, Wetzel CH, Buchalla W, Proescholdt M, Klein CA, Berneburg M, Schlitt HJ, Brabletz T, Ziegler C, Parkinson EK, Gaumann A, Geissler EK, Adamski J, Haferkamp S, Mycielska ME. Cancer-associated cells release citrate to support tumour metastatic progression. Life Sci Alliance 2021; 4:e202000903. [PMID: 33758075 PMCID: PMC7994318 DOI: 10.26508/lsa.202000903] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Citrate is important for lipid synthesis and epigenetic regulation in addition to ATP production. We have previously reported that cancer cells import extracellular citrate via the pmCiC transporter to support their metabolism. Here, we show for the first time that citrate is supplied to cancer by cancer-associated stroma (CAS) and also that citrate synthesis and release is one of the latter's major metabolic tasks. Citrate release from CAS is controlled by cancer cells through cross-cellular communication. The availability of citrate from CAS regulated the cytokine profile, metabolism and features of cellular invasion. Moreover, citrate released by CAS is involved in inducing cancer progression especially enhancing invasiveness and organ colonisation. In line with the in vitro observations, we show that depriving cancer cells of citrate using gluconate, a specific inhibitor of pmCiC, significantly reduced the growth and metastatic spread of human pancreatic cancer cells in vivo and muted stromal activation and angiogenesis. We conclude that citrate is supplied to tumour cells by CAS and citrate uptake plays a significant role in cancer metastatic progression.
Collapse
Affiliation(s)
- Konstantin Drexler
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | | | - Katrin Jordan
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Marianne Federlin
- Department of Conservative Dentistry and Periodontology, University Medical Center, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Christian Schmidl
- Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Gregor Madej
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Janina Tokarz
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Alexander Cecil
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Wolfgang Jagla
- Institute of Pathology, Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
- Center of Plastic, Aesthetic, Hand and Reconstructive Surgery, University of Regensburg, Regensburg, Germany
| | - Christine Wagner
- Department of Surgery, University Medical Center, Regensburg, Germany
| | | | - Stefanie Heinke
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Evan H Stanton
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Barbara Schwertner
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | - Dania Riegel
- Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Wolfgang Buchalla
- Department of Conservative Dentistry and Periodontology, University Medical Center, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Christoph A Klein
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Christine Ziegler
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Eric K Parkinson
- Centre for Immunobiology and Regenerative Medicine, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Andreas Gaumann
- Institute of Pathology, Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Edward K Geissler
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Munich, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Maria E Mycielska
- Department of Surgery, University Medical Center, Regensburg, Germany
| |
Collapse
|
39
|
Fan X, Zhou J, Yan X, Bi X, Liang J, Lu S, Luo L, Zhou D, Yin Z. Citrate activates autophagic death of prostate cancer cells via downregulation CaMKII/AKT/mTOR pathway. Life Sci 2021; 275:119355. [PMID: 33744326 DOI: 10.1016/j.lfs.2021.119355] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/08/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022]
Abstract
AIM The aim of this study was to explore the antitumor effect of citrate on prostate cancer and its underlying mechanism. MAIN METHODS CCK-8 and Colony formation assay were performed to detect the anti-proliferative effect of citrate on prostate cancer. Flow cytometry analysis was conducted to investigate the pro-apoptosis effect of citrate on prostate cancer. Immunofluorescence assay was taken to detect whether citrate induced autophagy in prostate cancer. Western blot and Immunohistochemical assay were performed to explore the underlying mechanism by which citrate activates autophagic death in prostate cancer cells. Xenograft tumorigenicity assay was conducted to explore whether citrate suppressed the growth of xenograft prostate tumors in vivo. KEY FINDINGS We found citrate could significantly induce apoptosis and autophagy of prostate cancer cells in vitro and in vivo. Furthermore, treatment with autophagy inhibitor (chloroquine) drastically suppresses the apoptosis rate of prostate cancer induced by citrate. Based on the Ca2+-chelating property of citrate, the further study suggested that citrate activates autophagic cell death in prostate cancer cells via downregulation CaMKII/AKT/mTOR pathway. Finally, citrate suppresses the growth of xenograft prostate tumors without remarkable toxicity in mice. SIGNIFICANCE Our study elucidated a novel molecular mechanism about the anti-cancer activities of citrate. That citrate activates autophagic cell death of prostate cancer via downregulation CaMKII/AKT/mTOR pathway and without remarkable toxicity in mice. This study suggests that citrate might be a promising therapeutic agent for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xirui Fan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Xintong Yan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaowen Bi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Juanjuan Liang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Shuai Lu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, People's Republic of China.
| | - Da Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 200032 Shanghai, People's Republic of China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
40
|
Coleman PS, Parlo RA. Warburg's Ghost-Cancer's Self-Sustaining Phenotype: The Aberrant Carbon Flux in Cholesterol-Enriched Tumor Mitochondria via Deregulated Cholesterogenesis. Front Cell Dev Biol 2021; 9:626316. [PMID: 33777935 PMCID: PMC7994618 DOI: 10.3389/fcell.2021.626316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
Interpreting connections between the multiple networks of cell metabolism is indispensable for understanding how cells maintain homeostasis or transform into the decontrolled proliferation phenotype of cancer. Situated at a critical metabolic intersection, citrate, derived via glycolysis, serves as either a combustible fuel for aerobic mitochondrial bioenergetics or as a continuously replenished cytosolic carbon source for lipid biosynthesis, an essentially anaerobic process. Therein lies the paradox: under what conditions do cells control the metabolic route by which they process citrate? The Warburg effect exposes essentially the same dilemma—why do cancer cells, despite an abundance of oxygen needed for energy-generating mitochondrial respiration with citrate as fuel, avoid catabolizing mitochondrial citrate and instead rely upon accelerated glycolysis to support their energy requirements? This review details the genesis and consequences of the metabolic paradigm of a “truncated” Krebs/TCA cycle. Abundant data are presented for substrate utilization and membrane cholesterol enrichment in tumors that are consistent with criteria of the Warburg effect. From healthy cellular homeostasis to the uncontrolled proliferation of tumors, metabolic alterations center upon the loss of regulation of the cholesterol biosynthetic pathway. Deregulated tumor cholesterogenesis at the HMGR locus, generating enhanced carbon flux through the cholesterol synthesis pathway, is an absolute prerequisite for DNA synthesis and cell division. Therefore, expedited citrate efflux from cholesterol-enriched tumor mitochondria via the CTP/SLC25A1 citrate transporter is fundamental for sustaining the constant demand for cytosolic citrate that fuels the elevated flow of carbons from acetyl-CoA through the deregulated pathway of cholesterol biosynthesis.
Collapse
Affiliation(s)
| | - Risa A Parlo
- Kingsborough Community College, Brooklyn, NY, United States
| |
Collapse
|
41
|
Muthusamy S, Zhu D, Rajalakshmi K, Zhu W, Wang S, Lee KB, Zhao L. Successive Detection of Zinc Ion and Citrate Using a Schiff Base Chemosensor for Enhanced Prostate Cancer Diagnosis in Biosystems. ACS APPLIED BIO MATERIALS 2021; 4:1932-1941. [PMID: 35014462 DOI: 10.1021/acsabm.0c01568] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sensitive and quantitative detection of prostate cancer (PC) requires a chemosensor with an applicable sensing strategy. A star-shaped Schiff base triaminoguanidine-integrated thiophene fluorophore TAT was rationally designed with nitrogen and sulfur atoms to coordinate with Zn2+ as the initial step and to chelate with citrate as the following step. Formation of the complex TAT-Zn2+ induced an intramolecular charge transfer and caused a red-shifted, Zn2+ concentration-dependent fluorescence at 507 nm. Chelation of TAT-Zn2+ with citrate led to an emission band at 692 nm upon an aggregation-induced emission mechanism. The distinctive fluorescence emissions of Zn2+ and citrate biomarkers were demonstrated first in on-site paper-based test strips showing gradually enhanced colors at yellow and red channels and second in both in vitro and in vivo by using PC3 cells and BALB/c nude mouse animal models, respectively. The in vitro test confirmed the mitochondria organelle-targeting property of TAT, and the in vivo performance manifested the successful application of the probe in recognizing the prostate cancer. This is the first applicable chemosensor that could be in continuous recognition of dual PC biomarkers Zn2+ and citrate in cancer diagnosis with a mitochondria organelle-targeting ability.
Collapse
Affiliation(s)
- Selvaraj Muthusamy
- Department of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Dongwei Zhu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Kanagaraj Rajalakshmi
- Department of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Weihua Zhu
- Department of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212013, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Kang-Bong Lee
- National Agenda Research Division, Korea Institute of Science & Technology, Hwarang-ro 14-gil 5 Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Long Zhao
- Department of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
42
|
The Mitochondrial Citrate Carrier SLC25A1/CIC and the Fundamental Role of Citrate in Cancer, Inflammation and Beyond. Biomolecules 2021; 11:biom11020141. [PMID: 33499062 PMCID: PMC7912299 DOI: 10.3390/biom11020141] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
The mitochondrial citrate/isocitrate carrier, CIC, has been shown to play an important role in a growing list of human diseases. CIC belongs to a large family of nuclear-encoded mitochondrial transporters that serve the fundamental function of allowing the transit of ions and metabolites through the impermeable mitochondrial membrane. Citrate is central to mitochondrial metabolism and respiration and plays fundamental activities in the cytosol, serving as a metabolic substrate, an allosteric enzymatic regulator and, as the source of Acetyl-Coenzyme A, also as an epigenetic modifier. In this review, we highlight the complexity of the mechanisms of action of this transporter, describing its involvement in human diseases and the therapeutic opportunities for targeting its activity in several pathological conditions.
Collapse
|
43
|
Zheng M, Wang W, Liu J, Zhang X, Zhang R. Lipid Metabolism in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:49-69. [PMID: 33740243 DOI: 10.1007/978-981-33-6785-2_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Metabolic reprogramming is one of the most critical hallmarks in cancer cells. In the past decades, mounting evidence has demonstrated that, besides the Warburg Effect, lipid metabolism dysregulation is also one of the essential characteristics of cancer cell metabolism. Lipids are water-insoluble molecules with diverse categories of phosphoglycerides, triacylglycerides, sphingolipids, sterols, etc. As the major utilization for energy storage, fatty acids are the primary building blocks for synthesizing triacylglycerides. And phosphoglycerides, sphingolipids, and sterols are the main components constructing biological membranes. More importantly, lipids play essential roles in signal transduction by functioning as second messengers or hormones. Much evidence has shown specific alterations of lipid metabolism in cancer cells. Consequently, the structural configuration of biological membranes, the energy homeostasis under nutrient stress, and the abundance of lipids in the intracellular signal transduction are affected by these alterations. Furthermore, lipid droplets accumulate in cancer cells and function adaptively to different types of harmful stress. This chapter reviews the regulation, functions, and therapeutic benefits of targeting lipid metabolism in cancer cells. Overall, this chapter highlights the significance of exploring more potential therapeutic strategies for malignant diseases by unscrambling lipid metabolism regulation in cancer cells.
Collapse
Affiliation(s)
- Minhua Zheng
- Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Wei Wang
- Department of Immunology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiao Zhang
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Rui Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, People's Republic of China.
- Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
44
|
Haferkamp S, Drexler K, Federlin M, Schlitt HJ, Berneburg M, Adamski J, Gaumann A, Geissler EK, Ganapathy V, Parkinson EK, Mycielska ME. Extracellular Citrate Fuels Cancer Cell Metabolism and Growth. Front Cell Dev Biol 2020; 8:602476. [PMID: 33425906 PMCID: PMC7793864 DOI: 10.3389/fcell.2020.602476] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer cells need excess energy and essential nutrients/metabolites not only to divide and proliferate but also to migrate and invade distant organs for metastasis. Fatty acid and cholesterol synthesis, considered a hallmark of cancer for anabolism and membrane biogenesis, requires citrate. We review here potential pathways in which citrate is synthesized and/or supplied to cancer cells and the impact of extracellular citrate on cancer cell metabolism and growth. Cancer cells employ different mechanisms to support mitochondrial activity and citrate synthesis when some of the necessary substrates are missing in the extracellular space. We also discuss the different transport mechanisms available for the entry of extracellular citrate into cancer cells and how citrate as a master metabolite enhances ATP production and fuels anabolic pathways. The available literature suggests that cancer cells show an increased metabolic flexibility with which they tackle changing environmental conditions, a phenomenon crucial for cancer cell proliferation and metastasis.
Collapse
Affiliation(s)
| | - Konstantin Drexler
- Department of Dermatology, University Medical Center, Regensburg, Germany
| | - Marianne Federlin
- Department of Conservative Dentistry and Periodontology, University Medical Center, Regensburg, Germany
| | - Hans J. Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Center, Regensburg, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Munich, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andreas Gaumann
- Institute of Pathology, Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Edward K. Geissler
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - E. Kenneth Parkinson
- Center for Immunobiology and Regenerative Medicine, Barts and The London School of Medicine and Dentistry, Blizard Institute, London, United Kingdom
| | - Maria E. Mycielska
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
45
|
Petillo A, Abruzzese V, Koshal P, Ostuni A, Bisaccia F. Extracellular Citrate Is a Trojan Horse for Cancer Cells. Front Mol Biosci 2020; 7:593866. [PMID: 33282912 PMCID: PMC7688668 DOI: 10.3389/fmolb.2020.593866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
The first intermediate in the mitochondrial tricarboxylic acid (TCA) cycle is citrate, which is essential and acts as a metabolic regulator for glycolysis, TCA cycle, gluconeogenesis, and fatty acid synthesis. Within the cytosol, citrate is cleaved by ATP citrate lyase (ACLY) into oxaloacetate (OAA) and acetyl-CoA; OAA can be used for neoglucogenesis or in the TCA cycle, while acetyl-CoA is the precursor of some biosynthetic processes, including the synthesis of fatty acids. Accumulating evidence suggests that citrate is involved in numerous physiological and pathophysiological processes such as inflammation, insulin secretion, neurological disorders, and cancer. Considering the crucial role of citrate to supply the acetyl-CoA pool for fatty acid synthesis and histone acetylation in tumors, in this study we evaluated the effect of citrate added to the growth medium on lipid deposition and histone H4 acetylation in hepatoma cells (HepG2). At low concentration, citrate increased both histone H4 acetylation and lipid deposition; at high concentration, citrate inhibited both, thus suggesting a crucial role of acetyl-CoA availability, which prompted us to investigate the effect of citrate on ACLY. In HepG2 cells, the expression of ACLY is correlated with histone acetylation, which, in turn, depends on citrate concentration. A decrease in H4 acetylation was also observed when citrate was added at a high concentration to immortalized human hepatic cells, whereas ACLY expression was unaffected, indicating a lack of control by histone acetylation. Considering the strong demand for acetyl-CoA but not for OAA in tumor cells, the exogenous citrate would behave like a trojan horse that carries OAA inside the cells and reduces ACLY expression and cellular metabolism. In addition, this study confirmed the already reported dual role of citrate both as a promoter of cell proliferation (at lower concentrations) and as an anticancer agent (at higher concentrations), providing useful tips on the use of citrate for the treatment of tumors.
Collapse
Affiliation(s)
- Agata Petillo
- Laboratory of Cell Biochemistry, Department of Sciences, University of Basilicata, Potenza, Italy
| | - Vittorio Abruzzese
- Laboratory of Cell Biochemistry, Department of Sciences, University of Basilicata, Potenza, Italy
| | - Prashant Koshal
- Laboratory of Cell Biochemistry, Department of Sciences, University of Basilicata, Potenza, Italy
| | - Angela Ostuni
- Laboratory of Cell Biochemistry, Department of Sciences, University of Basilicata, Potenza, Italy
| | - Faustino Bisaccia
- Laboratory of Cell Biochemistry, Department of Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
46
|
99mTc-citrate-gold nanoparticles as a tumor tracer: synthesis, characterization, radiolabeling and in-vivo studies. RADIOCHIM ACTA 2020. [DOI: 10.1515/ract-2019-3208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Targeted drug delivery system can reduce the side effects of high drug concentration by improving drug pharmacokinetics at lower doses. Citrate-gold nanoparticles (AuNPs) as a drug delivery system were synthesized via green nanotechnology technique to be used as a new imaging platform for tumor targeting. Citrate-AuNPs were synthesized with core size of 10 nm. Citrate-AuNPs were labeled with technetium-99m (99mTc) with radiochemical yield of 95.20 ± 2.70% with good in-vitro stability in both saline and human serum and well in-vivo studied in both normal and solid tumor bearing mice. The in-vivo biodistribution study of [99mTc]Tc-citrate-AuNPs in solid tumor bearing mice (as preliminary study) showed a high accumulation in tumor site with tumor/muscle of 4.35 ± 0.22 after 30 min post injection. The direct intratumoral (I.T) injection of [99mTc]Tc-citrate-AuNPs showed that this complex was retained in the tumor up to 77.86 ± 1.90 % at 5 min and still around 50.00 ± 1.42 % after 30 min post injection (p.i.). The newly presented nano-platform could be presented as a new potential radiopharmaceutical tumor imaging probe.
Collapse
|
47
|
Kampa JM, Kellner U, Marsching C, Ramallo Guevara C, Knappe UJ, Sahin M, Giampà M, Niehaus K, Bednarz H. Glioblastoma multiforme: Metabolic differences to peritumoral tissue and
IDH
‐mutated gliomas revealed by mass spectrometry imaging. Neuropathology 2020; 40:546-558. [DOI: 10.1111/neup.12671] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/20/2020] [Accepted: 03/22/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Judith M. Kampa
- Proteome and Metabolome Research, Faculty of Biology & Center for Biotechnology Bielefeld University Bielefeld Germany
| | - Udo Kellner
- Institut für Pathologie, Johannes Wesling Klinikum Minden Germany
| | - Christian Marsching
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS) Mannheim University of Applied Sciences Mannheim Germany
| | - Carina Ramallo Guevara
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS) Mannheim University of Applied Sciences Mannheim Germany
| | - Ulrich J. Knappe
- Klinik für Neurochirurgie, Johannes Wesling Klinikum Minden Germany
| | - Mikail Sahin
- Proteome and Metabolome Research, Faculty of Biology & Center for Biotechnology Bielefeld University Bielefeld Germany
| | - Marco Giampà
- Proteome and Metabolome Research, Faculty of Biology & Center for Biotechnology Bielefeld University Bielefeld Germany
| | - Karsten Niehaus
- Proteome and Metabolome Research, Faculty of Biology & Center for Biotechnology Bielefeld University Bielefeld Germany
| | - Hanna Bednarz
- Proteome and Metabolome Research, Faculty of Biology & Center for Biotechnology Bielefeld University Bielefeld Germany
| |
Collapse
|
48
|
Szeri F, Lundkvist S, Donnelly S, Engelke UFH, Rhee K, Williams CJ, Sundberg JP, Wevers RA, Tomlinson RE, Jansen RS, van de Wetering K. The membrane protein ANKH is crucial for bone mechanical performance by mediating cellular export of citrate and ATP. PLoS Genet 2020; 16:e1008884. [PMID: 32639996 PMCID: PMC7371198 DOI: 10.1371/journal.pgen.1008884] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/20/2020] [Accepted: 05/25/2020] [Indexed: 01/23/2023] Open
Abstract
The membrane protein ANKH was known to prevent pathological mineralization of joints and was thought to export pyrophosphate (PPi) from cells. This did not explain, however, the presence of ANKH in tissues, such as brain, blood vessels and muscle. We now report that in cultured cells ANKH exports ATP, rather than PPi, and, unexpectedly, also citrate as a prominent metabolite. The extracellular ATP is rapidly converted into PPi, explaining the role of ANKH in preventing ankylosis. Mice lacking functional Ank (Ankank/ank mice) had plasma citrate concentrations that were 65% lower than those detected in wild type control animals. Consequently, citrate excretion via the urine was substantially reduced in Ankank/ank mice. Citrate was even undetectable in the urine of a human patient lacking functional ANKH. The hydroxyapatite of Ankank/ank mice contained dramatically reduced levels of both, citrate and PPi and displayed diminished strength. Our results show that ANKH is a critical contributor to extracellular citrate and PPi homeostasis and profoundly affects bone matrix composition and, consequently, bone quality.
Collapse
Affiliation(s)
- Flora Szeri
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and PXE International Center of Excellence in Research and Clinical Care, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Stefan Lundkvist
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and PXE International Center of Excellence in Research and Clinical Care, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sylvia Donnelly
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and PXE International Center of Excellence in Research and Clinical Care, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Udo F. H. Engelke
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kyu Rhee
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Charlene J. Williams
- Cooper Medical School of Rowan University, Camden, New Jersey, United States of America
| | - John P. Sundberg
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Ron A. Wevers
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ryan E. Tomlinson
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Robert S. Jansen
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Koen van de Wetering
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and PXE International Center of Excellence in Research and Clinical Care, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
49
|
Parri M, Ippolito L, Cirri P, Ramazzotti M, Chiarugi P. Metabolic cell communication within tumour microenvironment: models, methods and perspectives. Curr Opin Biotechnol 2020; 63:210-219. [PMID: 32416546 DOI: 10.1016/j.copbio.2020.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/19/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023]
Abstract
Environmental cues are essential in defining tumour malignancy, by promoting tumour initiation, progression and metastatic spreading. Stromal cells may metabolically cooperate or compete with cancer cells, playing a mandatory role in defining cancer metabolic plasticity, potentially dictating the final tumour outcome. Assessing shared nutrients between different tumoural or stromal compartments is essential to understand the impact of environmental nutrients on the metabolic plasticity of tumours. Here, we review analytical and computational approaches for studying the tumour metabolic microenvironment, the destiny of nutrients shared among tumour and stromal populations, as well as the molecular modules of these metabolic relationships.
Collapse
Affiliation(s)
- M Parri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - L Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - P Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - M Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - P Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
50
|
Läsche M, Emons G, Gründker C. Shedding New Light on Cancer Metabolism: A Metabolic Tightrope Between Life and Death. Front Oncol 2020; 10:409. [PMID: 32300553 PMCID: PMC7145406 DOI: 10.3389/fonc.2020.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
Since the earliest findings of Otto Warburg, who discovered the first metabolic differences between lactate production of cancer cells and non-malignant tissues in the 1920s, much time has passed. He explained the increased lactate levels with dysfunctional mitochondria and aerobic glycolysis despite adequate oxygenation. Meanwhile, we came to know that mitochondria remain instead functional in cancer cells; hence, metabolic drift, rather than being linked to dysfunctional mitochondria, was found to be an active act of direct response of cancer cells to cell proliferation and survival signals. This metabolic drift begins with the use of sugars and the full oxidative phosphorylation via the mitochondrial respiratory chain to form CO2, and it then leads to the formation of lactic acid via partial oxidation. In addition to oncogene-driven metabolic reprogramming, the oncometabolites themselves alter cell signaling and are responsible for differentiation and metastasis of cancer cells. The aberrant metabolism is now considered a major characteristic of cancer within the past 15 years. However, the proliferating anabolic growth of a tumor and its spread to distal sites of the body is not explainable by altered glucose metabolism alone. Since a tumor consists of malignant cells and its tumor microenvironment, it was important for us to understand the bilateral interactions between the primary tumor and its microenvironment and the processes underlying its successful metastasis. We here describe the main metabolic pathways and their implications in tumor progression and metastasis. We also portray that metabolic flexibility determines the fate of the cancer cell and ultimately the patient. This flexibility must be taken into account when deciding on a therapy, since singular cancer therapies only shift the metabolism to a different alternative path and create resistance to the medication used. As with Otto Warburg in his days, we primarily focused on the metabolism of mitochondria when dealing with this scientific question.
Collapse
Affiliation(s)
- Matthias Läsche
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medicine Göttingen, Göttingen, Germany
| |
Collapse
|