1
|
Khan G, Hussain MS, Ahmad S, Alam N, Ali MS, Alam P. Metabolomics as a tool for understanding and treating triple-negative breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04234-4. [PMID: 40314763 DOI: 10.1007/s00210-025-04234-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous variant of breast cancer distinguished by a lack of targeted therapies, posing significant challenges in diagnosis and treatment. Metabolomics, the comprehensive study of small compounds in biological systems, has been identified as an instrument for revealing the metabolic underpinnings of TNBC. This review highlights recent advancements in metabolomic approaches, such as mass spectrometry and nuclear magnetic resonance, which have identified metabolic vulnerabilities, resistance mechanisms, and potential therapeutic targets. Key findings include alterations in fatty acid, amino acid, and glutathione metabolism, along with hypoxia-driven metabolic reprogramming that contributes to disease progression. The combination of metabolomics with multi-omics techniques, supported by advanced computational methods such as machine learning, offers a pathway to overcome challenges in data standardization and biological complexity. Emerging strategies, including the use of artificial intelligence and multidimensional omics approaches, are paving the way for personalized medicine by enabling the discovery of novel biomarkers and targeted therapies. Despite these advances, significant hurdles remain, including the need for robust data standardization, validation of findings in diverse patient cohorts, and seamless integration with clinical workflows. By addressing these challenges, metabolomics has the potential to revolutionize TNBC management, offering tools for early detection, precision therapy, and improved patient outcomes. This review underscores the importance of interdisciplinary collaboration to translate metabolomic insights into actionable clinical applications.
Collapse
Affiliation(s)
- Gyas Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand, 248007, India.
| | - Sarfaraz Ahmad
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Nawazish Alam
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Md Sajid Ali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| |
Collapse
|
2
|
Venkatesh D, Sarkar S, Kandasamy T, Ghosh SS. In-silico identification and validation of Silibinin as a dual inhibitor for ENO1 and GLUT4 to curtail EMT signaling and TNBC progression. Comput Biol Chem 2025; 115:108312. [PMID: 39689434 DOI: 10.1016/j.compbiolchem.2024.108312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
The aberrant metabolic reprogramming endows TNBC cells with sufficient ATP and lactate required for survival and metastasis. Hence, the intervention of the metabolic network represents a promising avenue to alleviate the Warburg effect in TNBC cells to impair their invasive and metastatic potential. Multitudinous in-silico analysis identified Enolase1 (ENO1) and the surface transporter protein, GLUT4 to be the potential targets for the abrogation of the metabolic network. The expression profiles of ENO1 and GLUT4 genes showed anomalous expression in various cancers, including breast cancer. Subsequently, the functional and physiological interactions of the target proteins were analyzed from the protein-protein interaction network. The pathway enrichment analysis identified the prime cancer signaling pathways in which these proteins are involved. Further, docking results bestowed Silibinin as the concurrent inhibitor of ENO1 and GLUT4. Moreover, the stable interaction of Silibinin with both proteins deciphered the binding free energies values of -48.86 and -104.31 KJ/mol from MMPBSA analysis and MD simulation, respectively. Furthermore, the cell viability, ROS assay, and live-dead imaging underscored the pronounced cytotoxicity of Silibinin, illuminating its capacity to incur apoptosis within TNBC cells. Additionally, glycolysis assay and gene expression analysis demonstrated the silibinin-mediated inhibition of the glycolysis pathway. Eventually, a lipidomic reprogramming towards fatty acid metabolism was established from the elevated lipid droplet accumulation, exogenous fatty acid uptake and de-novo lipogenesis. Nevertheless, repression of EMT and Wnt pathway progression by Silibinin was perceived from the gene expression studies. Overall, the current study highlights the tweaking of intricate signaling crosstalk between glycolysis and the Wnt pathway in TNBC cells through inhibiting ENO1 and GLUT4.
Collapse
Affiliation(s)
- Dheepika Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India.
| |
Collapse
|
3
|
Goncalves MD, Iyengar NM. Fueling metabolic disruption via FMD to boost chemotherapy in TNBC. Cell Metab 2025; 37:305-306. [PMID: 39908983 DOI: 10.1016/j.cmet.2024.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is highly glycolytic and lacks effective biomarkers for therapy response. The BREAKFAST trial showed that a fasting-mimicking diet (FMD) improved pathological complete response (pCR) rates to 56.6% compared to historical chemotherapy averages (30%-40%), with minimal severe adverse events. FMD's metabolic and immune-modulating effects warrant further study with immunotherapy.
Collapse
Affiliation(s)
| | - Neil M Iyengar
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
4
|
Martínez-Ramírez JM, Carmona C, Ramírez-Expósito MJ, Martínez-Martos JM. Extracting Knowledge from Machine Learning Models to Diagnose Breast Cancer. Life (Basel) 2025; 15:211. [PMID: 40003620 PMCID: PMC11856414 DOI: 10.3390/life15020211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
This study explored the application of explainable machine learning models to enhance breast cancer diagnosis using serum biomarkers, contrary to many studies that focus on medical images and demographic data. The primary objective was to develop models that are not only accurate but also provide insights into the factors driving predictions, addressing the need for trustworthy AI in healthcare. Several classification models were evaluated, including OneR, JRIP, the FURIA, J48, the ADTree, and the Random Forest, all of which are known for their explainability. The dataset included a variety of biomarkers, such as electrolytes, metal ions, marker proteins, enzymes, lipid profiles, peptide hormones, steroid hormones, and hormone receptors. The Random Forest model achieved the highest accuracy at 99.401%, followed closely by JRIP, the FURIA, and the ADTree at 98.802%. OneR and J48 achieved 98.204% accuracy. Notably, the models identified oxytocin as a key predictive biomarker, with most models featuring it in their rules. Other significant parameters included GnRH, β-endorphin, vasopressin, IRAP, and APB, as well as factors like iron, cholinesterase, the total protein, progesterone, 5-nucleotidase, and the BMI, which are considered clinically relevant to breast cancer pathogenesis. This study discusses the roles of the identified parameters in cancer development, thus underscoring the potential of explainable machine learning models for enhancing early breast cancer diagnosis by focusing on explainability and the use of serum biomarkers.The combination of both can lead to improved early detection and personalized treatments, emphasizing the potential of these methods in clinical settings. The identified markers also provide additional research and therapeutic targets for breast cancer pathogenesis and a deep understanding of their interactions, advancing personalized approaches to breast cancer management.
Collapse
Affiliation(s)
| | - Cristobal Carmona
- Department of Computer Science, University of Jaén, E-23071 Jaén, Spain; (J.M.M.-R.); (C.C.)
- Andalusian Research Institute in Data Science and Computational Intelligence, DASCI, University of Jaén, E-23071 Jaén, Spain
- Leicester School of Pharmacy, DeMontfort University, Leicester LE1 7RH, UK
| | - María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group CVI-1039, Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain;
| | - José Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group CVI-1039, Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain;
| |
Collapse
|
5
|
Ronghe R, Tavares AAS. The skeleton: an overlooked regulator of systemic glucose metabolism in cancer? Front Oncol 2024; 14:1481241. [PMID: 39588310 PMCID: PMC11586348 DOI: 10.3389/fonc.2024.1481241] [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: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Recent discoveries demonstrated the skeleton's role as an endocrine organ regulating whole-body glucose homeostasis. Glucose metabolism is critical for rapid cell proliferation and tumour growth through increasing glucose uptake and fermentation of glucose to lactate despite being in an aerobic environment. This hypothesis paper discusses emerging evidence on how bones can regulate whole-body glucose homeostasis with potential to impact on tumour growth and proliferation. Moreover, it proposes a clinical link between bone glucose metabolism and prognosis of cancer based on recent clinical trial data. Targeting metabolic pathways related with classic glucose metabolism and also bone metabolism, novel methods of cancer therapy and treatment could be developed. This paper objective is to highlight the need for future research on this altered metabolism with potential to change future management of cancer patients.
Collapse
Affiliation(s)
- Rucha Ronghe
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, United Kingdom
- Edinburgh Imaging, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Hu Q, Wang Y, Mao W. Knockdown of Glycolysis-Related LINC01070 Inhibits the Progression of Breast Cancer. Cureus 2024; 16:e60093. [PMID: 38860098 PMCID: PMC11163994 DOI: 10.7759/cureus.60093] [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] [Accepted: 05/11/2024] [Indexed: 06/12/2024] Open
Abstract
Accumulative evidence confirms that glycolysis and long non-coding RNAs (lncRNAs) are closely associated with tumor development. The aim of this study was to construct a novel prognostic model based on glycolysis-related lncRNAs (GRLs) in breast cancer patients. By performing Pearson correlation analysis and Lasso regression analysis on differentially expressed genes and lncRNAs associated with glycolysis in the Cancer Genome Atlas (TCGA) and Gene Set Enrichment Analysis (GSEA) datasets, we identified nine GRLs and constructed associated prognostic risk signature. Kaplan-Meier survival analysis and univariate and multivariate Cox analysis showed that patients in the low-risk group had a better prognosis. The receiver operator characteristics (ROC) curves showed that the area under the curve (AUC) of the prognostic risk signature predicting patients' overall survival at 1-, 3- and 5- years was 0.78, 0.71, and 0.71, respectively. Moreover, the validation curves also showed that the signature had better diagnostic efficacy and clinical predictive power. Furthermore, clone formation assay, EdU assay, and Transwell assay showed that knockdown of LINC01070 inhibited breast cancer progression. We developed a prognostic risk-associated GRLs signature that can accurately predict the breast cancer patient's prognostic status, and LINC01070 can be used as a potential biomarker for the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Qiang Hu
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| | - Yiduo Wang
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| | - Weipu Mao
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| |
Collapse
|
7
|
Zou J, Mai C, Lin Z, Zhou J, Lai G. Targeting metabolism of breast cancer and its implications in T cell immunotherapy. Front Immunol 2024; 15:1381970. [PMID: 38680483 PMCID: PMC11045902 DOI: 10.3389/fimmu.2024.1381970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Breast cancer is a prominent health issue amongst women around the world. Immunotherapies including tumor targeted antibodies, adoptive T cell therapy, vaccines, and immune checkpoint blockers have rejuvenated the clinical management of breast cancer, but the prognosis of patients remains dismal. Metabolic reprogramming and immune escape are two important mechanisms supporting the progression of breast cancer. The deprivation uptake of nutrients (such as glucose, amino acid, and lipid) by breast cancer cells has a significant impact on tumor growth and microenvironment remodeling. In recent years, in-depth researches on the mechanism of metabolic reprogramming and immune escape have been extensively conducted, and targeting metabolic reprogramming has been proposed as a new therapeutic strategy for breast cancer. This article reviews the abnormal metabolism of breast cancer cells and its impact on the anti-tumor activity of T cells, and further explores the possibility of targeting metabolism as a therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Jialuo Zou
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Cunjun Mai
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhiqin Lin
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jian Zhou
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Guie Lai
- Department of Breast Disease Comprehensive Center, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
8
|
Ahmed SBM, Radwan N, Amer S, Saheb Sharif-Askari N, Mahdami A, Samara KA, Halwani R, Jelinek HF. Assessing the Link between Diabetic Metabolic Dysregulation and Breast Cancer Progression. Int J Mol Sci 2023; 24:11816. [PMID: 37511575 PMCID: PMC10380477 DOI: 10.3390/ijms241411816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetes mellitus is a burdensome disease that affects various cellular functions through altered glucose metabolism. Several reports have linked diabetes to cancer development; however, the exact molecular mechanism of how diabetes-related traits contribute to cancer progression is not fully understood. The current study aimed to explore the molecular mechanism underlying the potential effect of hyperglycemia combined with hyperinsulinemia on the progression of breast cancer cells. To this end, gene dysregulation induced by the exposure of MCF7 breast cancer cells to hyperglycemia (HG), or a combination of hyperglycemia and hyperinsulinemia (HGI), was analyzed using a microarray gene expression assay. Hyperglycemia combined with hyperinsulinemia induced differential expression of 45 genes (greater than or equal to two-fold), which were not shared by other treatments. On the other hand, in silico analysis performed using a publicly available dataset (GEO: GSE150586) revealed differential upregulation of 15 genes in the breast tumor tissues of diabetic patients with breast cancer when compared with breast cancer patients with no diabetes. SLC26A11, ALDH1A3, MED20, PABPC4 and SCP2 were among the top upregulated genes in both microarray data and the in silico analysis. In conclusion, hyperglycemia combined with hyperinsulinemia caused a likely unique signature that contributes to acquiring more carcinogenic traits. Indeed, these findings might potentially add emphasis on how monitoring diabetes-related metabolic alteration as an adjunct to diabetes therapy is important in improving breast cancer outcomes. However, further detailed studies are required to decipher the role of the highlighted genes, in this study, in the pathogenesis of breast cancer in patients with a different glycemic index.
Collapse
Affiliation(s)
- Samrein B M Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Health, Wellbeing and Life Sciences, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Nada Radwan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sara Amer
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amena Mahdami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Kamel A Samara
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rabih Halwani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Herbert F Jelinek
- Department of Biomedical Engineering and Health Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
9
|
Oh M, Batty S, Banerjee N, Kim TH. High extracellular glucose promotes cell motility by modulating cell deformability and contractility via the cAMP-RhoA-ROCK axis in human breast cancer cells. Mol Biol Cell 2023; 34:ar79. [PMID: 37195739 PMCID: PMC10398875 DOI: 10.1091/mbc.e22-12-0560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
The mechanical properties, or mechanotypes, of cells are largely determined by their deformability and contractility. The ability of cancer cells to deform and generate contractile force is critical in multiple steps of metastasis. Identifying soluble cues that regulate cancer cell mechanotypes and understanding the underlying molecular mechanisms regulating these cellular mechanotypes could provide novel therapeutic targets to prevent metastasis. Although a strong correlation between high glucose level and cancer metastasis has been demonstrated, the causality has not been elucidated, and the underlying molecular mechanisms remain largely unknown. In this study, using novel high-throughput mechanotyping assays, we show that human breast cancer cells become less deformable and more contractile with increased extracellular glucose levels (>5 mM). These altered cell mechanotypes are due to increased F-actin rearrangement and nonmuscle myosin II (NMII) activity. We identify the cAMP-RhoA-ROCK-NMII axis as playing a major role in regulating cell mechanotypes at high extracellular glucose levels, whereas calcium and myosin light-chain kinase (MLCK) are not required. The altered mechanotypes are also associated with increased cell migration and invasion. Our study identifies key components in breast cancer cells that convert high extracellular glucose levels into changes in cellular mechanotype and behavior relevant in cancer metastasis.
Collapse
Affiliation(s)
- Mijung Oh
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Skylar Batty
- Undergraduate Pipeline Network Summer Research Program, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721
| | - Nayan Banerjee
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India
| | - Tae-Hyung Kim
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131
| |
Collapse
|
10
|
Munkácsy G, Santarpia L, Győrffy B. Therapeutic Potential of Tumor Metabolic Reprogramming in Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:ijms24086945. [PMID: 37108109 PMCID: PMC10138520 DOI: 10.3390/ijms24086945] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with clinical features of high metastatic potential, susceptibility to relapse, and poor prognosis. TNBC lacks the expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). It is characterized by genomic and transcriptional heterogeneity and a tumor microenvironment (TME) with the presence of high levels of stromal tumor-infiltrating lymphocytes (TILs), immunogenicity, and an important immunosuppressive landscape. Recent evidence suggests that metabolic changes in the TME play a key role in molding tumor development by impacting the stromal and immune cell fractions, TME composition, and activation. Hence, a complex inter-talk between metabolic and TME signaling in TNBC exists, highlighting the possibility of uncovering and investigating novel therapeutic targets. A better understanding of the interaction between the TME and tumor cells, and the underlying molecular mechanisms of cell-cell communication signaling, may uncover additional targets for better therapeutic strategies in TNBC treatment. In this review, we aim to discuss the mechanisms in tumor metabolic reprogramming, linking these changes to potential targetable molecular mechanisms to generate new, physical science-inspired clinical translational insights for the cure of TNBC.
Collapse
Affiliation(s)
- Gyöngyi Munkácsy
- National Laboratory for Drug Research and Development, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary
- Oncology Biomarker Research Group, Research Centre for Natural Sciences, Institute of Enzymology, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tűzoltó u. 5-7, 1094 Budapest, Hungary
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 5-7, 1094 Budapest, Hungary
| |
Collapse
|
11
|
Wu D, Yuan X, Zhou R, Chen W, Li W, Li Z, Li X, Zhu R, Wang H, Yang Y. Aqueous extract of Sanghuangporus baumii induces autophagy to inhibit cervical carcinoma growth. Food Funct 2023; 14:2374-2384. [PMID: 36779533 DOI: 10.1039/d2fo02887e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Sanghuangporus baumii, an edible fungus rich in heteropolysaccharides, has been found to have some anti-cervical cancer effects. In the current study, the effects of an aqueous extract of S. baumii on cervical cancer were investigated in a U14 cervical carcinoma cell implanted female Kunming mouse model. An aqueous extract of S. baumii (SHWE) was administered to tumor-bearing mice by gavage for 21 days. SHWE treatment significantly inhibited tumor growth by 67.4% at a dose of 400 mg per kg bodyweight. Transcriptomic results showed that the expression of key genes GABARAP, VMP1, VAMP8 and STX17 which are involved in the autophagy pathway was regulated after SHWE treatment, suggesting that SHWE may induce autophagy in tumors. The results were further confirmed by measuring the LC3II/LC3I ratio using western blotting. Moreover, some differentially expressed genes were involved in the insulin signaling pathway, implying that SHWE induced autophagy by disturbing glucose uptake and utilization in tumors. The analysis of the gut microbiota indicated that SHWE treatment stimulated the proliferation of Akkermansia, a well-known probiotic that presented benefits in metabolic regulation and cancer therapy. In conclusion, SHWE administration modified the gut microbiota, disturbed the glucose metabolism and induced autophagy in tumors, and then inhibited the development of cervical carcinoma in vivo.
Collapse
Affiliation(s)
- Di Wu
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Xuemei Yuan
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Ruijie Zhou
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Wanchao Chen
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Wen Li
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Zhengpeng Li
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| | - Xueyin Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Rui Zhu
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hualin Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China.
| | - Yan Yang
- Institute of Edible Fungi, Shanghai Academy of Agriculture Sciences, National Engineering Research Center of Edible Fungi, Key Laboratory of Edible Fungi Resources and Utilization (South), Ministry of Agriculture, Shanghai 201403, China.
| |
Collapse
|
12
|
Novel Gene Signatures Promote Epithelial-Mesenchymal Transition (EMT) in Glucose Deprivation-Based Microenvironment to Predict Recurrence-Free Survival in Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2023; 2023:6114976. [PMID: 36866237 PMCID: PMC9974289 DOI: 10.1155/2023/6114976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Background Current research studies have suggested that glucose deprivation (GD)-based tumor microenvironment (TME) can promote epithelial-mesenchymal transition (EMT) of tumor cells, leading to tumor invasion and metastasis. However, no one has yet studied detailedly the synthetic studies that include GD features in TME with EMT status. In our research, we comprehensively developed and validated a robust signature regarding GD and EMT status to provide prognostic value for patients with liver cancer. Methods GD and EMT status were estimated with transcriptomic profiles based on WGCNA and t-SNE algorithms. Two cohorts of training (TCGA_LIHC) and validation (GSE76427) datasets were analyzed with the Cox regression and logistic regression analyses. We identified a 2-mRNA signature to establish a GD-EMT-based gene risk model for the prediction of HCC relapse. Results Patients with significant GD-EMT status were divided into two subgroups: GDlow/EMTlow and GDhigh/EMThigh, with the latter having significantly worse recurrence-free survival (P < 0.01). We employed the least absolute shrinkage and selection operator (LASSO) technique as a method for HNF4A and SLC2A4 filtering and constructing a risk score for risk stratification. In the multivariate analysis, this risk score predicted recurrence-free survival (RFS) in both the discovery and validation cohorts and remained valid in patients stratified by TNM stage and age at diagnosis. The nomogram that combines risk score and TNM stage as well as age produces improved performance and net benefits in the analysis of calibration and decision curves in training and validation groups. Conclusions The GD-EMT-based signature predictive model may provide a prognosis classifier for HCC patients with a high risk of postoperative recurrence to decrease the relapse rate.
Collapse
|
13
|
Shum HCE, Wu K, Vadgama J, Wu Y. Potential Therapies Targeting the Metabolic Reprogramming of Diabetes-Associated Breast Cancer. J Pers Med 2023; 13:157. [PMID: 36675817 PMCID: PMC9861470 DOI: 10.3390/jpm13010157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
In recent years, diabetes-associated breast cancer has become a significant clinical challenge. Diabetes is not only a risk factor for breast cancer but also worsens its prognosis. Patients with diabetes usually show hyperglycemia and hyperinsulinemia, which are accompanied by different glucose, protein, and lipid metabolism disorders. Metabolic abnormalities observed in diabetes can induce the occurrence and development of breast cancer. The changes in substrate availability and hormone environment not only create a favorable metabolic environment for tumorigenesis but also induce metabolic reprogramming events required for breast cancer cell transformation. Metabolic reprogramming is the basis for the development, swift proliferation, and survival of cancer cells. Metabolism must also be reprogrammed to support the energy requirements of the biosynthetic processes in cancer cells. In addition, metabolic reprogramming is essential to enable cancer cells to overcome apoptosis signals and promote invasion and metastasis. This review aims to describe the major metabolic changes in diabetes and outline how cancer cells can use cellular metabolic changes to drive abnormal growth and proliferation. We will specifically examine the mechanism of metabolic reprogramming by which diabetes may promote the development of breast cancer, focusing on the role of glucose metabolism, amino acid metabolism, and lipid metabolism in this process and potential therapeutic targets. Although diabetes-associated breast cancer has always been a common health problem, research focused on finding treatments suitable for the specific needs of patients with concurrent conditions is still limited. Most studies are still currently in the pre-clinical stage and mainly focus on reprogramming the glucose metabolism. More research targeting the amino acid and lipid metabolism is needed.
Collapse
Affiliation(s)
- Hang Chee Erin Shum
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ke Wu
- David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1748 E. 118th Street, Los Angeles, CA 90095, USA
| | - Jaydutt Vadgama
- David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1748 E. 118th Street, Los Angeles, CA 90095, USA
| | - Yong Wu
- David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1748 E. 118th Street, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Ren C, Wang Q, Wang S, Zhou H, Xu M, Li H, Li Y, Chen X, Liu X. Metabolic syndrome-related prognostic index: Predicting biochemical recurrence and differentiating between cold and hot tumors in prostate cancer. Front Endocrinol (Lausanne) 2023; 14:1148117. [PMID: 37033267 PMCID: PMC10080042 DOI: 10.3389/fendo.2023.1148117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND The prostate, as an endocrine and reproductive organ, undergoes complex hormonal and metabolic changes. Recent studies have shown a potential relationship between metabolic syndrome and the progression and recurrence of prostate cancer (PCa). This study aimed to construct a metabolic syndrome-related prognostic index (MSRPI) to predict biochemical recurrence-free survival (BFS) in patients with PCa and to identify cold and hot tumors to improve individualized treatment for patients with PCa. METHODS The Cancer Genome Atlas database provided training and test data, and the Gene Expression Omnibus database provided validation data. We extracted prognostic differentially expressed metabolic syndrome-related genes (DEMSRGs) related to BFS using univariate Cox analysis and identified potential tumor subtypes by consensus clustering. The least absolute shrinkage and selection operator (LASSO) algorithm and multivariate Cox regression were used to construct the MSRPI. We further validated the predictive power of the MSRPI using KaplanMeier survival analysis and receiver operating characteristic (ROC) curves, both internally and externally. Drug sensitivity was predicted using the half-maximal inhibitory concentration (IC50). Finally, we explored the landscape of somatic mutations in the risk groups. RESULTS Forty-six prognostic DEMSRGs and two metabolic syndrome-associated molecular clusters were identified. Cluster 2 was more immunogenic. Seven metabolic syndrome-related genes (CSF3R, TMEM132A, STAB1, VIM, DUOXA1, PILRB, and SLC2A4) were used to construct risk equations. The high-risk index was significantly associated with a poor BFS, which was also validated in the validation cohort. The area under the ROC curve (AUC) for BFS at 1-, 3-, and 5- year in the entire cohort was 0.819, 0.785, and 0.772, respectively, demonstrating the excellent predictive power of the MSRPI. Additionally, the MSRPI was found to be an independent prognostic factor for BFS in PCa. More importantly, MSRPI helped differentiate between cold and hot tumors. Hot tumors were associated with the high-risk group. Multiple drugs demonstrated significantly lower IC50 values in the high-risk group, offering the prospect of precision therapy for patients with PCa. CONCLUSION The MSRPI developed in this study was able to predict biochemical recurrence in patients with PCa and identify cold and hot tumors. MSRPI has the potential to improve personalized precision treatment.
Collapse
Affiliation(s)
- Congzhe Ren
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qihua Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Zhou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingming Xu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hu Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Urology, Shanxian Central Hospital (Affiliated Huxi Hospital of Jining Medical University), Heze, China
| | - Yuezheng Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangyu Chen
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Xiaoqiang Liu,
| |
Collapse
|
15
|
Cerebrolysin Alleviating Effect on Glutamate-Mediated Neuroinflammation Via Glutamate Transporters and Oxidative Stress. J Mol Neurosci 2022; 72:2292-2302. [PMID: 36333611 DOI: 10.1007/s12031-022-02078-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Glutamate, one of the most important excitatory neurotransmitters, acts as a signal transducer in peripheral tissues and endocrine cells. Excessive glutamate secretion has been shown to cause excitotoxicity and neurodegenerative disease. Cerebrolysin is a mixture of enzymatically treated peptides derived from pig brain including neurotrophic factors, like brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and ciliary neurotrophic factor (CNTF). The present study investigated the protective effects of cerebrolysin on glutamate transporters (EAAT 1, EAAT 2) and cytokines (IL-1β and IL-10) activity in glutamate-mediated neurotoxicity. Primary cortex neuron culture was exposed to glutamate and successively treated with various cerebrolysin concentrations for 24 and 48 h. Our data showed that cerebrolysin primarily protects neurons by decreasing glutamate concentration in the synaptic cleft. In addition, Cerebrolysin can decrease oxidative stress and neuron cell damage by increasing antioxidant activity and decreasing inflammation cytokine levels.
Collapse
|
16
|
Prasad M, Veeraraghavan VP, Jayaraman S. Tumorigenic potential of GLUT4: A therapeutic target for head and neck squamous cell carcinoma. Oral Oncol 2022; 133:106061. [PMID: 35952585 DOI: 10.1016/j.oraloncology.2022.106061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Monisha Prasad
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India.
| |
Collapse
|
17
|
Abd El-Azeem MA, Ali MA, El-Shorbagy SH. Expression of GLUT4 and FAP in urothelial bladder carcinoma: correlation with angiogenesis and clinicopathological characteristics. J Egypt Natl Canc Inst 2022; 34:40. [DOI: 10.1186/s43046-022-00145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/27/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Urothelial carcinoma (UC) is the most common type of bladder cancer. Glucose transporter 4 (GLUT4) is one of glucose transporter proteins’ family which facilitates glucose transport inside the cells. It was found to be overexpressed in several malignant tumors. Cancer-associated fibroblasts (CAFs) are heterogeneous stromal cells located adjacent to cancer cells and are considered one of the most important tumor stromal cells. They have been associated with enhancing tumor growth and invasion. GLUT4 expression in malignant epithelial cells and fibroblast activation protein (FAP) expression in CAFs of UC in relation to angiogenesis and clinicopathological characteristics are studied in this work.
Materials and methods
The study was carried out on 72 paraffin blocks of UC (27 radical cystectomies and 45 transurethral resections). Immunohistochemical staining was performed with GLUT4, FAP, and CD34 antibodies. Expression of GLUT4 and FAP was classified according to the staining intensities and percentages into low and high groups. CD34-stained microvessels’ mean count in five microscopic fields (×200) was taken as the microvessel density (MVD).
Results
GLUT4 overexpression was detected in 32 UC. It was significantly associated with high-grade tumors, advanced primary tumor (pT) stage, lymphovascular invasion (LVI), and regional lymph node invasion. High FAP expression was appreciated in 27 UC and was significantly linked to LVI and advanced TNM staging. Intratumor MVD significantly increased in UC with muscle invasion, LVI, and regional lymph node and/or distant metastasis. A significant positive correlation between GLUT4, FAP expression, and MVD was found.
Conclusion
GLUT4 and FAP expression was significantly associated with increased intratumor MVD and adverse clinicopathological factors.
Collapse
|
18
|
ACBD3 Bioinformatic Analysis and Protein Expression in Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms23168881. [PMID: 36012147 PMCID: PMC9408326 DOI: 10.3390/ijms23168881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
ACBD3 overexpression has previously been found to correlate with worse prognosis for breast cancer patients and, as an incredibly diverse protein in both function and cellular localisation, ACBD3 may have a larger role in breast cancer than previously thought. This study further investigated ACBD3′s role in breast cancer. Bioinformatic databases were queried to characterise ACBD3 expression and mutation in breast cancer and to investigate how overexpression affects breast cancer patient outcomes. Immunohistochemistry was carried out to examine ACBD3 location within cells and tissue structures. ACBD3 was more highly expressed in breast cancer than in any other cancer or matched normal tissue, and expression over the median level resulted in reduced relapse-free, overall, and distant metastasis-free survival for breast cancer patients as a whole, with some differences observed between subtypes. IHC analysis found that ACBD3 levels varied based on hormone receptor status, indicating that ACBD3 could be a candidate biomarker for poor patient prognosis in breast cancer and may possibly be a biomarker for ER signal reprogramming of precancerous breast tissue.
Collapse
|
19
|
Rossi V, Govoni M, Farabegoli F, Di Stefano G. Lactate is a potential promoter of tamoxifen resistance in MCF7 cells. Biochim Biophys Acta Gen Subj 2022; 1866:130185. [PMID: 35661802 DOI: 10.1016/j.bbagen.2022.130185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Tamoxifen is a widely used estrogen receptor inhibitor, whose clinical success is limited by the development of acquired resistance. This compound was also found to inhibit mitochondrial function, causing increased glycolysis and lactate production. Lactate has been widely recognized as a signaling molecule, showing the potential of modifying gene expression. These metabolic effects of tamoxifen can by hypothesized to contribute in driving drug resistance. METHODS To test this hypothesis, we used MCF7 cells together with a tamoxifen resistant cell line (MCF7-TAM). Experiments were aimed at verifying whether enhanced lactate exposure can affect the phenotype of MCF7 cells, conferring them features mirroring those observed in the tamoxifen resistant culture. RESULTS The obtained results suggested that enhanced lactate in MCF7 cells medium can increase the expression of tafazzin (TAZ) and telomerase complex (TERC, TERT) genes, reducing the cells' attitude to undergo senescence. In long term lactate-exposed cells, signs of EGFR activation, a pathway related to acquired tamoxifen resistance, was also observed. CONCLUSIONS The obtained results suggested lactate as a potential promoter of tamoxifen resistance. The off-target effects of this compound could play a role in hindering its therapeutic efficacy. GENERAL SIGNIFICANCE The features of acquired tamoxifen resistance have been widely characterized at the molecular level; in spite of their heterogeneity, poorly responsive cells were often found to display upregulated glycolysis. Our results suggest that this metabolic asset is not simply a result of neoplastic progression, but can play an active part in driving this process.
Collapse
Affiliation(s)
- Valentina Rossi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Marzia Govoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Fulvia Farabegoli
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Giuseppina Di Stefano
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy.
| |
Collapse
|
20
|
Barbier C, Mansour A, Ismailova A, Sarmadi F, Scarlata DA, Bouttier M, Zeitouni C, Wang C, Gleason JL, White JH. Molecular mechanisms of bifunctional vitamin D receptor agonist-histone deacetylase inhibitor hybrid molecules in triple-negative breast cancer. Sci Rep 2022; 12:6745. [PMID: 35468986 PMCID: PMC9038752 DOI: 10.1038/s41598-022-10740-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
The active form of vitamin D, 1,25-dihydroxyvitamin D (1,25D), and its analogues signal through the nuclear vitamin D receptor (VDR), a ligand-regulated transcription factor, and have been extensively investigated as anticancer agents. 1,25D and its analogs have potential in combination therapies because they exhibit synergistic activities with other anticancer agents such as histone deacetylase inhibitors (HDACi). We have developed a series of hybrid molecules that combine HDACi within the backbone of a VDR agonist and thus represent fully integrated bifunctional molecules. They exhibit anti-tumor efficacy in reducing tumor growth and metastases in an aggressive model of triple-negative breast cancer. However, their solubility is limited by their hydrophobic diarylpentane cores. Our goals here were two-fold: (1) to improve the solubility of hybrids by introducing nitrogen into diarylpentane cores, and (2) to investigate the molecular mechanisms underlying their anti-tumor efficacy by performing comparative gene expression profiling studies with 1,25D and the potent HDACi suberoylanilide hydroxamic acid (SAHA). We found that substituting aryl with pyrydyl rings did not sacrifice bifunctionality and modestly improved solubility. Notably, one compound, AM-193, displayed enhanced potency as a VDR agonist and in cellular assays of cytotoxicity. RNAseq studies in triple negative breast cancer cells revealed that gene expression profiles of hybrids were very similar to that of 1,25D, as was that observed with 1,25D and SAHA combined. The effects of SAHA alone on gene expression were limited and distinct from those 1,25D or hybrids. The combined results suggest that efficacy of hybrids arises from targeting HDACs that do not have a direct role in gene regulation. Moreover, pathways analysis revealed that hybrids regulate numerous genes controlling immune cell infiltration into tumors and suppress the expression of several secreted molecules that promote breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Camille Barbier
- Departments of Physiology, McGill University, Montreal, QC, Canada
| | - Ali Mansour
- Departments of Chemistry, McGill University, Montreal, QC, Canada
| | - Aiten Ismailova
- Departments of Physiology, McGill University, Montreal, QC, Canada
| | - Fatemeh Sarmadi
- Departments of Physiology, McGill University, Montreal, QC, Canada
| | - David A Scarlata
- Departments of Chemistry, McGill University, Montreal, QC, Canada
| | | | - Camille Zeitouni
- Departments of Physiology, McGill University, Montreal, QC, Canada
| | - Catherine Wang
- Departments of Physiology, McGill University, Montreal, QC, Canada
| | - James L Gleason
- Departments of Chemistry, McGill University, Montreal, QC, Canada.
| | - John H White
- Departments of Physiology, McGill University, Montreal, QC, Canada.
- Departments of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Suteau V, Bukasa-Kakamba J, Virjogh-Cenciu B, Adenis A, Sabbah N, Drak Alsibai K. Pathological Significance of GLUT-1 Expression in Breast Cancer Cells in Diabetic and Obese Patients: The French Guiana Study. Cancers (Basel) 2022; 14:cancers14020437. [PMID: 35053598 PMCID: PMC8774256 DOI: 10.3390/cancers14020437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 01/12/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary This study describes the clinical, histological, and molecular features of breast cancer in French Guiana, and characterizes the expression of the tumor metabolic marker GLUT-1 in breast cancers cells in diabetic and obese patients compared to a control group. This study reveals an overall overexpression of GLUT-1 in 60% of invasive breast carcinomas and in all medullary pattern and carcinoma in situ lesions. Our results highlight the potential role of GLUT-1 as a tumor metabolic prognostic marker and also as an interesting target therapy, independently of patient metabolic disorder. Abstract The prevalence of obesity and type 2 diabetes is higher in French Guiana compared to mainland France. These metabolic disorders are associated with an increased risk of cancer. One of the factors involved is hyperinsulinemia that promotes the action of glucose transporter 1 (GLUT-1). The objective of this study is to characterize the expression of GLUT-1 in breast cancers cells in diabetic and obese patients compared to those who are not and to describe the clinical and histological prognostic factors of breast cancer in this population. We conducted a monocentric study including patients with breast cancer diagnosed between 2014 and 2020. Patients were classified into three groups: diabetes, obesity, and control group. The GLUT-1 expression was assessed by immunohistochemistry. In total, 199 patients were included in this study. The median age was 53.5 years, and the median tumor size was 2.8 cm. Luminal A was the most frequent molecular type (58.1%), followed by the triple-negative type (19.9%). The breast cancer in our population was characterized by a younger age at diagnosis, more aggressive molecular types, and larger tumor size. Thus, we suggest the advancement of the age of breast cancer screening in this territory. A total of 144 patients (31 diabetes, 22 obese, and 91 control group) were included for the study of GLUT-1 expression. Overexpression of GLUT-1 was observed in 60.4% of cases and in all carcinoma in situ lesions. GLUT-1 overexpression was associated with more aggressive cancers. This overexpression is correlated with high histological grade, high proliferation index, and aggressive molecular types. Our study found no difference in GLUT-1 expression between the diabetic or obese patients and the control group. These results highlight the potential role of GLUT-1 as a tumor metabolic prognostic marker and also as an interesting target therapy, independently of patient metabolic disorder.
Collapse
Affiliation(s)
- Valentin Suteau
- Department of Pathology, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - John Bukasa-Kakamba
- Department of Endocrinology and Metabolic Diseases, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana; (J.B.-K.); (N.S.)
| | - Beatrice Virjogh-Cenciu
- Department of Medicine, Hôpital de jour Adults, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - Antoine Adenis
- Clinical Investigation Center Antilles French Guiana (CIC INSERM 1424), Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - Nadia Sabbah
- Department of Endocrinology and Metabolic Diseases, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana; (J.B.-K.); (N.S.)
| | - Kinan Drak Alsibai
- Department of Pathology, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
- Center of Biological Resources (CRB Amazonie), Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana
- Correspondence: ; Tel.: +594-594395231
| |
Collapse
|
22
|
Wegner MS, Schömel N, Olzomer EM, Trautmann S, Olesch C, Byrne FL, Brüne B, Gurke R, Ferreirós N, Weigert A, Geisslinger G, Hoehn KL. Increased glucosylceramide production leads to decreased cell energy metabolism and lowered tumor marker expression in non-cancerous liver cells. Cell Mol Life Sci 2021; 78:7025-7041. [PMID: 34626204 PMCID: PMC8558193 DOI: 10.1007/s00018-021-03958-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most difficult cancer types to treat. Liver cancer is often diagnosed at late stages and therapeutic treatment is frequently accompanied by development of multidrug resistance. This leads to poor outcomes for cancer patients. Understanding the fundamental molecular mechanisms leading to liver cancer development is crucial for developing new therapeutic approaches, which are more efficient in treating cancer. Mice with a liver specific UDP-glucose ceramide glucosyltransferase (UGCG) knockout (KO) show delayed diethylnitrosamine (DEN)-induced liver tumor growth. Accordingly, the rationale for our study was to determine whether UGCG overexpression is sufficient to drive cancer phenotypes in liver cells. We investigated the effect of UGCG overexpression (OE) on normal murine liver (NMuLi) cells. Increased UGCG expression results in decreased mitochondrial respiration and glycolysis, which is reversible by treatment with EtDO-P4, an UGCG inhibitor. Furthermore, tumor markers such as FGF21 and EPCAM are lowered following UGCG OE, which could be related to glucosylceramide (GlcCer) and lactosylceramide (LacCer) accumulation in glycosphingolipid-enriched microdomains (GEMs) and subsequently altered signaling protein phosphorylation. These cellular processes lead to decreased proliferation in NMuLi/UGCG OE cells. Our data show that increased UGCG expression itself does not induce pro-cancerous processes in normal liver cells, which indicates that increased GlcCer expression leads to different outcomes in different cancer types.
Collapse
Affiliation(s)
- Marthe-Susanna Wegner
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany. .,School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Nina Schömel
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Ellen M Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sandra Trautmann
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Catherine Olesch
- Faculty of Medicine, Institute of Biochemistry I, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Robert Gurke
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Nerea Ferreirós
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, House 74, Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
23
|
Li H, Qi Z, Niu Y, Yang Y, Li M, Pang Y, Liu M, Cheng X, Xu M, Wang Z. FBP1 regulates proliferation, metastasis, and chemoresistance by participating in C-MYC/STAT3 signaling axis in ovarian cancer. Oncogene 2021; 40:5938-5949. [PMID: 34363022 PMCID: PMC8497274 DOI: 10.1038/s41388-021-01957-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/17/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Fructose-1,6-bisphosphatase (FBP1) is a rate-limiting enzyme in gluconeogenesis and an important tumor suppressor in human malignancies. Here, we aimed to investigate the expression profile of FBP1 in ovarian cancer, the molecular mechanisms that regulate FBP1 expression and to examine how the FBP1 regulatory axis contributes to tumorigenesis and progression in ovarian cancer. We showed that FBP1 expression was significantly decreased in ovarian cancer tissues compared with normal ovarian tissues, and low-FBP1 expression predicted poor prognosis in patients with ovarian cancer. The enhanced expression of FBP1 in ovarian cancer cell lines suppressed proliferation and 2-D/3-D invasion, reduced aerobic glycolysis, and sensitized cancer cells to cisplatin-induced apoptosis. Moreover, DNA methylation and C-MYC binding at the promoter inhibited FBP1 expression. Furthermore, through physical interactions with signal transducer and activator of transcription 3 (STAT3), FBP1 suppressed nuclear translocation of STAT3 and exerted its non-metabolic enzymatic activity to induce the dysfunction of STAT3. Thus, our study suggests that FBP1 may be a valuable prognostic predictor for ovarian cancer. C-MYC-dependent downregulation of FBP1 acted as a tumor suppressor via modulating STAT3, and the C-MYC/FBP1/STAT3 axis could be a therapeutic target.
Collapse
Affiliation(s)
- Haoran Li
- Cancer Institute and Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihao Qi
- Department of General Surgery, Shanghai First People's Hospital, Shanghai Jiaotong Univeristy School of Medicine, Shanghai, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Yufei Yang
- Cancer Institute and Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Clinical Medicine Transformation Center and Office of Academic Research, Shanghai Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengjiao Li
- Cancer Institute and Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yangyang Pang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Mingming Liu
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xi Cheng
- Cancer Institute and Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Midie Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Department of Pathology and Biobank, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Ziliang Wang
- Cancer Institute and Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Clinical Medicine Transformation Center and Office of Academic Research, Shanghai Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
24
|
Shi Z, Liu J, Wang F, Li Y. Integrated analysis of Solute carrier family-2 members reveals SLC2A4 as an independent favorable prognostic biomarker for breast cancer. Channels (Austin) 2021; 15:555-568. [PMID: 34488531 PMCID: PMC8425726 DOI: 10.1080/19336950.2021.1973788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Most of Solute carrier family-2 (SLC2) members play a key role of facilitative transporters, and glucose transporter (GLUT) proteins encoded by SLC2s can transport hexoses or polyols. However, the function and mechanism of SLC2s remain unclear in human cancers. Here, we explored the dysregulated expression, prognostic values, epigenetic, genetic alterations, and biomolecular network of SLC2s in human cancers. According to the data from public-omicsrepository, SLC2A4 (GLUT4) was found to be significantly downregulated in most cancers, and higher messenger RNA (mRNA) expression of SLC2A4 significantly associated with better prognosis of breast cancer (BRCA) patients. Moreover, DNA hypermethylation in the promoter of SLC2A4 may affect the regulation of its mRNA expression, and SLC2A4 was strongly correlated with pathways, including the translocation of SLC2A4 to the plasma membrane and PID INSULIN PATHWAY. In conclusion, these results provide insight into SLC2s in human cancers and suggest that SLC2A4 could be an unfavorable prognostic biomarker for the survival of BRCA patients.
Collapse
Affiliation(s)
- Zhenyu Shi
- Department of Predictive Medicine,Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, HenanUniversity,Kaifeng,China
| | - Jiahao Liu
- Department of Predictive Medicine,Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, HenanUniversity,Kaifeng,China
| | - Fei Wang
- Department of Predictive Medicine,Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, HenanUniversity,Kaifeng,China
| | - Yongqiang Li
- Department of Predictive Medicine,Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, HenanUniversity,Kaifeng,China
| |
Collapse
|
25
|
Zhang Q, Yang X, Liu H. Extracellular Vesicles in Cancer Metabolism: Implications for Cancer Diagnosis and Treatment. Technol Cancer Res Treat 2021; 20:15330338211037821. [PMID: 34427131 PMCID: PMC8388228 DOI: 10.1177/15330338211037821] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Metabolic reprogramming is one of the most common characteristics of cancer cells. The metabolic alterations of glucose, amino acids and lipids can support the aggressive phenotype of cancer cells. Exosomes, a kind of extracellular vesicles, participate in the intercellular communication through transferring bioactive molecules. Increasing evidence has demonstrated that enzymes, metabolites and non-coding RNAs in exosomes are responsible for the metabolic alteration of cancer cells. In this review, we summarize the past and recent findings of exosomes in altering cancer metabolism and elaborate on the role of the specific enzymes, metabolites and non-coding RNAs transferred by exosomes. Moreover, we give evidence of the role of exosomes in cancer diagnosis and treatment. Finally, we discuss the existing problems in the study and application of exosomes in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Pliszka M, Szablewski L. Glucose Transporters as a Target for Anticancer Therapy. Cancers (Basel) 2021; 13:cancers13164184. [PMID: 34439338 PMCID: PMC8394807 DOI: 10.3390/cancers13164184] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary For mammalian cells, glucose is a major source of energy. In the presence of oxygen, a complete breakdown of glucose generates 36 molecules of ATP from one molecule of glucose. Hypoxia is a hallmark of cancer; therefore, cancer cells prefer the process of glycolysis, which generates only two molecules of ATP from one molecule of glucose, and cancer cells need more molecules of glucose in comparison with normal cells. Increased uptake of glucose by cancer cells is due to increased expression of glucose transporters. However, overexpression of glucose transporters, promoting the process of carcinogenesis, and increasing aggressiveness and invasiveness of tumors, may have also a beneficial effect. For example, upregulation of glucose transporters is used in diagnostic techniques such as FDG-PET. Therapeutic inhibition of glucose transporters may be a method of treatment of cancer patients. On the other hand, upregulation of glucose transporters, which are used in radioiodine therapy, can help patients with cancers. Abstract Tumor growth causes cancer cells to become hypoxic. A hypoxic condition is a hallmark of cancer. Metabolism of cancer cells differs from metabolism of normal cells. Cancer cells prefer the process of glycolysis as a source of ATP. Process of glycolysis generates only two molecules of ATP per one molecule of glucose, whereas the complete oxidative breakdown of one molecule of glucose yields 36 molecules of ATP. Therefore, cancer cells need more molecules of glucose in comparison with normal cells. Increased uptake of glucose by these cells is due to overexpression of glucose transporters, especially GLUT1 and GLUT3, that are hypoxia responsive, as well as other glucose transport proteins. Increased expression of these carrier proteins may be used in anticancer therapy. This phenomenon is used in diagnostic techniques such as FDG-PET. It is also suggested, and there are observations, that therapeutic inhibition of glucose transporters may be a method in treatment of cancer patients. On the other hand, there are described cases, in which upregulation of glucose transporters, as, for example, NIS, which is used in radioiodine therapy, can help patients with cancer. The aim of this review is the presentation of possibilities, and how glucose transporters can be used in anticancer therapy.
Collapse
|
27
|
Akt Isoforms: A Family Affair in Breast Cancer. Cancers (Basel) 2021; 13:cancers13143445. [PMID: 34298660 PMCID: PMC8306188 DOI: 10.3390/cancers13143445] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Breast cancer is the second leading cause of cancer-related death in women in the United States. The Akt signaling pathway is deregulated in approximately 70% of patients with breast cancer. While targeting Akt is an effective therapeutic strategy for the treatment of breast cancer, there are several members in the Akt family that play distinct roles in breast cancer. However, the function of Akt isoforms depends on many factors. This review analyzes current progress on the isoform-specific functions of Akt isoforms in breast cancer. Abstract Akt, also known as protein kinase B (PKB), belongs to the AGC family of protein kinases. It acts downstream of the phosphatidylinositol 3-kinase (PI3K) and regulates diverse cellular processes, including cell proliferation, cell survival, metabolism, tumor growth and metastasis. The PI3K/Akt signaling pathway is frequently deregulated in breast cancer and plays an important role in the development and progression of breast cancer. There are three closely related members in the Akt family, namely Akt1(PKBα), Akt2(PKBβ) and Akt3(PKBγ). Although Akt isoforms share similar structures, they exhibit redundant, distinct as well as opposite functions. While the Akt signaling pathway is an important target for cancer therapy, an understanding of the isoform-specific function of Akt is critical to effectively target this pathway. However, our perception regarding how Akt isoforms contribute to the genesis and progression of breast cancer changes as we gain new knowledge. The purpose of this review article is to analyze current literatures on distinct functions of Akt isoforms in breast cancer.
Collapse
|
28
|
Son SW, Yun BD, Song MG, Lee JK, Choi SY, Kuh HJ, Park JK. The Hypoxia-Long Noncoding RNA Interaction in Solid Cancers. Int J Mol Sci 2021; 22:ijms22147261. [PMID: 34298879 PMCID: PMC8307739 DOI: 10.3390/ijms22147261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is one of the representative microenvironment features in cancer and is considered to be associated with the dismal prognosis of patients. Hypoxia-driven cellular pathways are largely regulated by hypoxia-inducible factors (HIFs) and notably exert influence on the hallmarks of cancer, such as stemness, angiogenesis, invasion, metastasis, and the resistance towards apoptotic cell death and therapeutic resistance; therefore, hypoxia has been considered as a potential hurdle for cancer therapy. Growing evidence has demonstrated that long noncoding RNAs (lncRNAs) are dysregulated in cancer and take part in gene regulatory networks owing to their various modes of action through interacting with proteins and microRNAs. In this review, we focus attention on the relationship between hypoxia/HIFs and lncRNAs, in company with the possibility of lncRNAs as candidate molecules for controlling cancer.
Collapse
Affiliation(s)
- Seung Wan Son
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
| | - Ba Da Yun
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
| | - Mun Gyu Song
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
| | - Jin Kyeong Lee
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
| | - Hyo Jeong Kuh
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jong Kook Park
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.W.S.); (B.D.Y.); (M.G.S.); (J.K.L.); (S.Y.C.)
- Correspondence: ; Tel.: +82-33-248-2114
| |
Collapse
|
29
|
Huang P, Zhu S, Liang X, Zhang Q, Luo X, Liu C, Song L. Regulatory Mechanisms of LncRNAs in Cancer Glycolysis: Facts and Perspectives. Cancer Manag Res 2021; 13:5317-5336. [PMID: 34262341 PMCID: PMC8275123 DOI: 10.2147/cmar.s314502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cells exhibit distinct metabolic characteristics that employ glycolysis to provide energy and intermediary metabolites. This aberrant metabolic phenotype favors cancer progression. LncRNAs are transcripts longer than 200 nucleotides that do not encode proteins. LncRNAs contribute to cancer progression and therapeutic resistance and affect aerobic glycolysis via multiple mechanisms, including modulating glycolytic transporters and enzymes. Further, dysregulated signaling pathways are vital for glycolysis. In this review, we highlight regulatory mechanisms for lncRNAs in aerobic glycolysis that provide novel insights into cancer development. Moreover, a comprehensive understanding of the regulatory mechanisms of lncRNAs in aerobic glycolysis can provide new strategies for clinical cancer management.
Collapse
Affiliation(s)
- Peng Huang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Shaomi Zhu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Xiaohong Luo
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China
| |
Collapse
|
30
|
Demény MA, Virág L. The PARP Enzyme Family and the Hallmarks of Cancer Part 1. Cell Intrinsic Hallmarks. Cancers (Basel) 2021; 13:cancers13092042. [PMID: 33922595 PMCID: PMC8122967 DOI: 10.3390/cancers13092042] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/02/2021] [Accepted: 04/20/2021] [Indexed: 12/21/2022] Open
Abstract
The 17-member poly (ADP-ribose) polymerase enzyme family, also known as the ADP-ribosyl transferase diphtheria toxin-like (ARTD) enzyme family, contains DNA damage-responsive and nonresponsive members. Only PARP1, 2, 5a, and 5b are capable of modifying their targets with poly ADP-ribose (PAR) polymers; the other PARP family members function as mono-ADP-ribosyl transferases. In the last decade, PARP1 has taken center stage in oncology treatments. New PARP inhibitors (PARPi) have been introduced for the targeted treatment of breast cancer 1 or 2 (BRCA1/2)-deficient ovarian and breast cancers, and this novel therapy represents the prototype of the synthetic lethality paradigm. Much less attention has been paid to other PARPs and their potential roles in cancer biology. In this review, we summarize the roles played by all PARP enzyme family members in six intrinsic hallmarks of cancer: uncontrolled proliferation, evasion of growth suppressors, cell death resistance, genome instability, reprogrammed energy metabolism, and escape from replicative senescence. In a companion paper, we will discuss the roles of PARP enzymes in cancer hallmarks related to cancer-host interactions, including angiogenesis, invasion and metastasis, evasion of the anticancer immune response, and tumor-promoting inflammation. While PARP1 is clearly involved in all ten cancer hallmarks, an increasing body of evidence supports the role of other PARPs in modifying these cancer hallmarks (e.g., PARP5a and 5b in replicative immortality and PARP2 in cancer metabolism). We also highlight controversies, open questions, and discuss prospects of recent developments related to the wide range of roles played by PARPs in cancer biology. Some of the summarized findings may explain resistance to PARPi therapy or highlight novel biological roles of PARPs that can be therapeutically exploited in novel anticancer treatment paradigms.
Collapse
Affiliation(s)
- Máté A. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (M.A.D.); (L.V.)
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- MTA-DE Cell Biology and Signaling Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (M.A.D.); (L.V.)
| |
Collapse
|
31
|
Echeverría C, Nualart F, Ferrada L, Smith GJ, Godoy AS. Hexose Transporters in Cancer: From Multifunctionality to Diagnosis and Therapy. Trends Endocrinol Metab 2021; 32:198-211. [PMID: 33518451 DOI: 10.1016/j.tem.2020.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022]
Abstract
Cancer cells increase their metabolic activity by enhancing glucose uptake through overexpression of hexose transporters (Gluts). Gluts also have the capacity to transport other molecules besides glucose, including fructose, mannose, and dehydroascorbic acid (DHA), the oxidized form of vitamin C. The majority of research studies in this field have focused on the role of glucose transport and metabolism in cancer, leaving a substantial gap in our knowledge of the contribution of other hexoses and DHA in cancer biology. Here, we summarize the most recent advances in understanding the role that the multifunctional transport capacity of Gluts plays in biological and clinical aspects of cancer, and how these characteristics can be exploited in the search for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Carolina Echeverría
- Centro de Biología Celular y Biomedicina, Universidad San Sebastián, Santiago, Chile; Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Francisco Nualart
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile; Centro de Microscopía Avanzada, Universidad de Concepción, Concepción, Chile
| | - Luciano Ferrada
- Centro de Microscopía Avanzada, Universidad de Concepción, Concepción, Chile
| | - Gary J Smith
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Alejandro S Godoy
- Centro de Biología Celular y Biomedicina, Universidad San Sebastián, Santiago, Chile; Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
32
|
Silva C, Andrade N, Guimarães JT, Patrício E, Martel F. The in vitro effect of the diabetes-associated markers insulin, leptin and oxidative stress on cellular characteristics promoting breast cancer progression is GLUT1-dependent. Eur J Pharmacol 2021; 898:173980. [PMID: 33647254 DOI: 10.1016/j.ejphar.2021.173980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 01/09/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) associate with increased incidence and mortality from many cancers, including breast cancer. The mechanisms involved in this relation remain poorly understood. Our study aimed to investigate the in vitro effect of high levels of glucose, insulin, leptin, TNF-α, INF-γ and oxidative stress (induced with tert-butylhydroperoxide (TBH)), which are associated with T2DM, upon glucose uptake by breast cancer (MCF-7 and MDA-MB-231) and non-cancer (MCF-12A) cells and to correlate this effect with their effects upon cellular characteristics associated with cancer progression (cell proliferation, viability, migration, angiogenesis and apoptosis). 3H-DG uptake was markedly inhibited by a selective GLUT1 inhibitor (BAY-876) in all cell lines, proving that 3H-DG uptake is mainly GLUT1-mediated. TBH (2.5 μM), insulin (50 nM), leptin (500 ng/ml) and INF-y (100 ng/ml) stimulate GLUT1-mediated 3H-DG (1 mM) uptake by both ER-positive and triple-negative breast cancer cell lines. TBH and leptin, but not insulin and INF-γ, increase GLUT1 mRNA levels. Insulin and leptin (in both ER-positive and triple-negative breast cancer cell lines) and TBH (in the triple-negative cell line) have a proproliferative effect and leptin possesses a cytoprotective effect in both breast cancer cell lines that can contribute to cancer progression. The effects of TBH, insulin, leptin and INF-γ upon breast cancer cell proliferation and viability are GLUT1-dependent. In conclusion, T2DM-associated characteristics induce changes in GLUT1-mediated glucose uptake that can contribute to cancer progression. Moreover, we conclude that BAY-876 can be a strong candidate for development of a new effective anticancer agent against breast cancer.
Collapse
Affiliation(s)
- Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal; REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - João Tiago Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal; Institute of Public Health, University of Porto, Porto, Portugal
| | - Emília Patrício
- Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
33
|
Yamada K, Nishimura T, Wakiya M, Satoh E, Fukuda T, Amaya K, Bando Y, Hirano H, Ishikawa T. Protein co-expression networks identified from HOT lesions of ER+HER2-Ki-67high luminal breast carcinomas. Sci Rep 2021; 11:1705. [PMID: 33462336 PMCID: PMC7814020 DOI: 10.1038/s41598-021-81509-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/05/2021] [Indexed: 01/20/2023] Open
Abstract
Patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative/Ki-67-high (ER+HER2-Ki-67high) luminal breast cancer have a worse prognosis and do not respond to hormonal treatment and chemotherapy. This study sought to identify disease-related protein networks significantly associated with this subtype, by assessing in-depth proteomes of 10 lesions of high and low Ki-67 values (HOT, five; COLD, five) microdissected from the five tumors. Weighted correlation network analysis screened by over-representative analysis identified the five modules significantly associated with the HOT lesions. Pathway enrichment analysis, together with causal network analysis, revealed pathways of ribosome-associated quality controls, heat shock response by oxidative stress and hypoxia, angiogenesis, and oxidative phosphorylation. A semi-quantitative correlation of key-protein expressions, protein co-regulation analysis, and multivariate correlation analysis suggested co-regulations via network-network interaction among the four HOT-characteristic modules. Predicted highly activated master and upstream regulators were most characteristic to ER-positive breast cancer and associated with oncogenic transformation, as well as resistance to chemotherapy and endocrine therapy. Interestingly, inhibited intervention causal networks of numerous chemical inhibitors were predicted within the top 10 lists for the WM2 and WM5 modules, suggesting involvement of potential therapeutic targets in those data-driven networks. Our findings may help develop therapeutic strategies to benefit patients.
Collapse
Affiliation(s)
- Kimito Yamada
- Department of Breast Surgery, Tokyo Medical University Hachioji Medical Centre, Tokyo, 193-0998, Japan
- Department of Breast Surgery, Tokyo Medical University Hospital, Tokyo, 160-0023, Japan
| | - Toshihide Nishimura
- Department of Translational Medicine Informatics, St. Marianna University School of Medicine, Kanagawa, 216-8511, Japan.
| | - Midori Wakiya
- Department of Diagnostic Pathology, Tokyo Medical University Hachioji Medical Centre, Tokyo, 193-0998, Japan
| | - Eiichi Satoh
- Department of Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Tetsuya Fukuda
- Research and Development, Biosys Technologies Inc, Tokyo, 152-0031, Japan
| | - Keigo Amaya
- Department of Breast Surgery, Tokyo Medical University Hachioji Medical Centre, Tokyo, 193-0998, Japan
| | - Yasuhiko Bando
- Research and Development, Biosys Technologies Inc, Tokyo, 152-0031, Japan
| | - Hiroshi Hirano
- Department of Diagnostic Pathology, Tokyo Medical University Hachioji Medical Centre, Tokyo, 193-0998, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery, Tokyo Medical University Hospital, Tokyo, 160-0023, Japan
| |
Collapse
|
34
|
Ramírez-Expósito MJ, Dueñas-Rodríguez B, Carrera-González MP, Navarro-Cecilia J, Martínez-Martos JM. Insulin-Regulated Aminopeptidase in Women with Breast Cancer: A Role beyond the Regulation of Oxytocin and Vasopressin. Cancers (Basel) 2020; 12:cancers12113252. [PMID: 33158090 PMCID: PMC7694176 DOI: 10.3390/cancers12113252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Insulin-regulated aminopeptidase (IRAP) is a well-known enzyme involved mainly in the regulation of the peptide hormones, oxytocin and vasopressin. However, this enzyme activity has hardly been analyzed in breast cancer patients. Additionally, the influence of both the hormonal status (pre or postmenopause) and the administration of neoadjuvant chemotherapy have rarely been studied. We show that there is a weak association between IRAP activity and the circulating levels of peptide hormones with variations depending on the hormonal status and the neoadjuvant treatment, and propose a role beyond oxytocin and vasopressin regulation that is related to the local mammary renin-angiotensin system and glucose transportation to the cells. Abstract Insulin-regulated aminopeptidase (IRAP) is the only enzyme known to cleave oxytocin and vasopressin; however, it is also the high-affinity binding site for angiotensin IV (AngIV) receptor type 4 (AT4) ligands and it is related to insulin-dependent glucose transporters through the translocation of the glucose transporter type 4 (GLUT4). Previous studies have demonstrated an association between IRAP activity and the number and size of mammary tumors in an animal model of breast cancer (BC). Also, a highly significant increase in IRAP activity has been found in BC tissue from women patients. Here, we found no changes in circulating IRAP in premenopausal (preMP) women, but it increased significantly in postmenopausal (postMP) women not treated with neoadjuvant chemotherapy (NACH). However, in women treated with NACH, IRAP activity increased in both preMP and postMP women. Two years of follow-up indicated lower levels of IRAP activity in untreated preMP women, but a return to control levels in untreated postMP women, while IRAP activity returned to control levels in women treated with NACH. Circulating oxytocin decreased in both preMP and postMP women during the follow-up period. Differences in Oxytocin appeared between preMP and postMP women treated with NACH, but not in women who were not treated with NACH. On the contrary, circulating vasopressin increased in untreated and treated preMP and postMP women, with most of the differences related to the hormonal status as well as the neoadjuvant treatment during the two year follow-up We propose that IRAP is involved in mechanisms related not only to oxytocin and/or vasopressin regulation, but also to the local mammary RAS through AngIV and its role in glucose transportation through the IRAP/GLUT4 system.
Collapse
Affiliation(s)
- María Jesús Ramírez-Expósito
- Experimental and Clinical Physiopathology Research Group, Department of Health Sciences, School of Experimental and Health Sciences, University of Jaén, E-23071 Jaén, Spain; (M.J.R.-E.); (B.D.-R.); (M.P.C.-G.); (J.N.-C.)
| | - Basilio Dueñas-Rodríguez
- Experimental and Clinical Physiopathology Research Group, Department of Health Sciences, School of Experimental and Health Sciences, University of Jaén, E-23071 Jaén, Spain; (M.J.R.-E.); (B.D.-R.); (M.P.C.-G.); (J.N.-C.)
- Unit of Breast Pathology, Complejo Hospitalario de Jaén, E-23007 Jaén, Spain
| | - María Pilar Carrera-González
- Experimental and Clinical Physiopathology Research Group, Department of Health Sciences, School of Experimental and Health Sciences, University of Jaén, E-23071 Jaén, Spain; (M.J.R.-E.); (B.D.-R.); (M.P.C.-G.); (J.N.-C.)
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and Nursing, Instituto Maimónides de Investigación Biomédica de Córdoba, University of Cordoba, 14004 Córdoba, Spain
| | - Joaquín Navarro-Cecilia
- Experimental and Clinical Physiopathology Research Group, Department of Health Sciences, School of Experimental and Health Sciences, University of Jaén, E-23071 Jaén, Spain; (M.J.R.-E.); (B.D.-R.); (M.P.C.-G.); (J.N.-C.)
- Unit of Breast Pathology, Complejo Hospitalario de Jaén, E-23007 Jaén, Spain
| | - Jose Manuel Martínez-Martos
- Experimental and Clinical Physiopathology Research Group, Department of Health Sciences, School of Experimental and Health Sciences, University of Jaén, E-23071 Jaén, Spain; (M.J.R.-E.); (B.D.-R.); (M.P.C.-G.); (J.N.-C.)
- Correspondence: ; Tel.: +34-953-212-600; Fax: +34-953-212-943
| |
Collapse
|
35
|
Harguindey S, Alfarouk K, Polo Orozco J, Fais S, Devesa J. Towards an Integral Therapeutic Protocol for Breast Cancer Based upon the New H +-Centered Anticancer Paradigm of the Late Post-Warburg Era. Int J Mol Sci 2020; 21:E7475. [PMID: 33050492 PMCID: PMC7589677 DOI: 10.3390/ijms21207475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Department of Pharmacology, Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
36
|
Let-7a-5p inhibits triple-negative breast tumor growth and metastasis through GLUT12-mediated warburg effect. Cancer Lett 2020; 495:53-65. [PMID: 32946964 DOI: 10.1016/j.canlet.2020.09.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is known for its aggressive phenotype with limited treatment modalities and poor prognosis. The Warburg effect (aerobic glycolysis) is a hallmark of cancer that serves as a promising target for diagnosis and therapy. However, how aerobic glycolysis regulates TNBC remains largely unknown. Here, we show that the glucose transporter (GLUT) family member GLUT12 promotes TNBC tumor growth and metastasis in vitro and in vivo through regulating aerobic glycolysis. MicroRNA let-7a-5p, a tumor suppressor, inhibited GLUT12 expression by targeting its 3'-untranslated region, and suppressed GLUT12-mediated TNBC tumor growth, metastasis, and glycolytic function, including alterations of glucose uptake, lactate production, ATP generation, extracellular acidification rate, and oxygen consumption rate. Inhibiting aerobic glycolysis abolished the ability of let-7a-5p and GLUT12 to regulate TNBC cell proliferation, migration and invasion. In TNBC patients, GLUT12 was significantly upregulated, and let-7a-5p expression was inversely correlated with GLUT12 expression. High expression of let-7a-5p and GLUT12 predicted better and worse clinical outcomes, respectively. Taken together, our results indicate that the let-7a-5p/GLUT12 axis plays key roles in TNBC tumor growth and metastasis, and aerobic glycolysis, and is a potential target for TNBC treatment.
Collapse
|
37
|
Gadaleta E, Fourgoux P, Pirró S, Thorn GJ, Nelan R, Ironside A, Rajeeve V, Cutillas PR, Lobley AE, Wang J, Gea E, Ross-Adams H, Bessant C, Lemoine NR, Jones LJ, Chelala C. Characterization of four subtypes in morphologically normal tissue excised proximal and distal to breast cancer. NPJ Breast Cancer 2020; 6:38. [PMID: 32885042 PMCID: PMC7442642 DOI: 10.1038/s41523-020-00182-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/06/2020] [Indexed: 01/20/2023] Open
Abstract
Widespread mammographic screening programs and improved self-monitoring allow for breast cancer to be detected earlier than ever before. Breast-conserving surgery is a successful treatment for select women. However, up to 40% of women develop local recurrence after surgery despite apparently tumor-free margins. This suggests that morphologically normal breast may harbor early alterations that contribute to increased risk of cancer recurrence. We conducted a comprehensive transcriptomic and proteomic analysis to characterize 57 fresh-frozen tissues from breast cancers and matched histologically normal tissues resected proximal to (<2 cm) and distant from (5-10 cm) the primary tumor, using tissues from cosmetic reduction mammoplasties as baseline. Four distinct transcriptomic subtypes are identified within matched normal tissues: metabolic; immune; matrisome/epithelial-mesenchymal transition, and non-coding enriched. Key components of the subtypes are supported by proteomic and tissue composition analyses. We find that the metabolic subtype is associated with poor prognosis (p < 0.001, HR6.1). Examination of genes representing the metabolic signature identifies several genes able to prognosticate outcome from histologically normal tissues. A subset of these have been reported for their predictive ability in cancer but, to the best of our knowledge, these have not been reported altered in matched normal tissues. This study takes an important first step toward characterizing matched normal tissues resected at pre-defined margins from the primary tumor. Unlocking the predictive potential of unexcised tissue could prove key to driving the realization of personalized medicine for breast cancer patients, allowing for more biologically-driven analyses of tissue margins than morphology alone.
Collapse
Affiliation(s)
- Emanuela Gadaleta
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Pauline Fourgoux
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
- Centre for Computational Biology, Life Sciences Initiative, Queen Mary University of London, London, EC1M 6BQ UK
| | - Stefano Pirró
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Graeme J. Thorn
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Rachel Nelan
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Alastair Ironside
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Vinothini Rajeeve
- Center for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Pedro R. Cutillas
- Center for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Anna E. Lobley
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Jun Wang
- Center for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Esteban Gea
- Centre for Computational Biology, Life Sciences Initiative, Queen Mary University of London, London, EC1M 6BQ UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS UK
| | - Helen Ross-Adams
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Conrad Bessant
- Centre for Computational Biology, Life Sciences Initiative, Queen Mary University of London, London, EC1M 6BQ UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, E1 4NS UK
| | - Nicholas R. Lemoine
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Louise J. Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
| | - Claude Chelala
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ UK
- Centre for Computational Biology, Life Sciences Initiative, Queen Mary University of London, London, EC1M 6BQ UK
| |
Collapse
|
38
|
Fernández-Calero T, Davyt M, Perelmuter K, Chalar C, Bampi G, Persson H, Tosar JP, Hafstað V, Naya H, Rovira C, Bollati-Fogolín M, Ehrlich R, Flouriot G, Ignatova Z, Marín M. Fine-tuning the metabolic rewiring and adaptation of translational machinery during an epithelial-mesenchymal transition in breast cancer cells. Cancer Metab 2020; 8:8. [PMID: 32699630 PMCID: PMC7368990 DOI: 10.1186/s40170-020-00216-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/26/2020] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT BACKGROUND During breast cancer progression, the epithelial to mesenchymal transition has been associated with metastasis and endocrine therapy resistance; however, the underlying mechanisms remain elusive. To gain insight into this process, we studied the transition undergone by MCF7-derived cells, which is driven by the constitutive nuclear expression of a MKL1 variant devoid of the actin-binding domain (MKL1 ΔN200). We characterized the adaptive changes that occur during the MKL1-induced cellular model and focused on regulation of translation machinery and metabolic adaptation. METHODS We performed a genome-wide analysis at the transcriptional and translational level using ribosome profiling complemented with RNA-Seq and analyzed the expression of components of the translation machinery and enzymes involved in energy metabolism. NGS data were correlated with metabolomic measurements and quantification of specific mRNAs extracted from polysomes and western blots. RESULTS Our results reveal the expression profiles of a luminal to basal-like state in accordance with an epithelial to mesenchymal transition. During the transition, the synthesis of ribosomal proteins and that of many translational factors was upregulated. This overexpression of the translational machinery appears to be regulated at the translational level. Our results indicate an increase of ribosome biogenesis and translation activity. We detected an extensive metabolic rewiring occurring in an already "Warburg-like" context, in which enzyme isoform switches and metabolic shunts indicate a crucial role of HIF-1α along with other master regulatory factors. Furthermore, we detected a decrease in the expression of enzymes involved in ribonucleotide synthesis from the pentose phosphate pathway. During this transition, cells increase in size, downregulate genes associated with proliferation, and strongly upregulate expression of cytoskeletal and extracellular matrix genes. CONCLUSIONS Our study reveals multiple regulatory events associated with metabolic and translational machinery adaptation during an epithelial mesenchymal-like transition process. During this major cellular transition, cells achieve a new homeostatic state ensuring their survival. This work shows that ribosome profiling complemented with RNA-Seq is a powerful approach to unveil in-depth global adaptive cellular responses and the interconnection among regulatory circuits, which will be helpful for identification of new therapeutic targets.
Collapse
Affiliation(s)
- Tamara Fernández-Calero
- Biochemistry-Molecular Biology Section, Faculty of Sciences, Universidad de la República, Iguá 4225, CP 11400 Montevideo, Uruguay
- Bioinformatics Unit, Institut Pasteur Montevideo, Mataojo, 2020 Montevideo, Uruguay
- Departamento de Ciencias Exactas y Naturales, Universidad Católica del Uruguay, Av. 8 de Octubre, 2738 Montevideo, Uruguay
| | - Marcos Davyt
- Biochemistry-Molecular Biology Section, Faculty of Sciences, Universidad de la República, Iguá 4225, CP 11400 Montevideo, Uruguay
| | - Karen Perelmuter
- Cell Biology Unit, Institut Pasteur Montevideo, Mataojo, 2020 Montevideo, Uruguay
| | - Cora Chalar
- Biochemistry-Molecular Biology Section, Faculty of Sciences, Universidad de la República, Iguá 4225, CP 11400 Montevideo, Uruguay
| | - Giovana Bampi
- Institute for Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Helena Persson
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University Cancer Center, Lund University, SE-223 63 Lund, Sweden
| | - Juan Pablo Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Mataojo, 2020 Montevideo, Uruguay
- Analytical Biochemistry Unit, Nuclear Research Center, Faculty of Science, Universidad de la República, Montevideo, Uruguay
| | - Völundur Hafstað
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University Cancer Center, Lund University, SE-223 63 Lund, Sweden
| | - Hugo Naya
- Bioinformatics Unit, Institut Pasteur Montevideo, Mataojo, 2020 Montevideo, Uruguay
| | - Carlos Rovira
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University Cancer Center, Lund University, SE-223 63 Lund, Sweden
| | | | - Ricardo Ehrlich
- Biochemistry-Molecular Biology Section, Faculty of Sciences, Universidad de la República, Iguá 4225, CP 11400 Montevideo, Uruguay
- Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gilles Flouriot
- Université de Rennes 1-IRSET, Campus Santé de Villejean, 35000 Rennes, France
| | - Zoya Ignatova
- Institute for Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Mónica Marín
- Biochemistry-Molecular Biology Section, Faculty of Sciences, Universidad de la República, Iguá 4225, CP 11400 Montevideo, Uruguay
| |
Collapse
|
39
|
Wang Z, Jiang Q, Dong C. Metabolic reprogramming in triple-negative breast cancer. Cancer Biol Med 2020; 17:44-59. [PMID: 32296576 PMCID: PMC7142847 DOI: 10.20892/j.issn.2095-3941.2019.0210] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/30/2019] [Indexed: 02/06/2023] Open
Abstract
Since triple-negative breast cancer (TNBC) was first defined over a decade ago, increasing studies have focused on its genetic and molecular characteristics. Patients diagnosed with TNBC, compared to those diagnosed with other breast cancer subtypes, have relatively poor outcomes due to high tumor aggressiveness and lack of targeted treatment. Metabolic reprogramming, an emerging hallmark of cancer, is hijacked by TNBC to fulfill bioenergetic and biosynthetic demands; maintain the redox balance; and further promote oncogenic signaling, cell proliferation, and metastasis. Understanding the mechanisms of metabolic remodeling may guide the design of metabolic strategies for the effective intervention of TNBC. Here, we review the metabolic reprogramming of glycolysis, oxidative phosphorylation, amino acid metabolism, lipid metabolism, and other branched pathways in TNBC and explore opportunities for new biomarkers, imaging modalities, and metabolically targeted therapies.
Collapse
Affiliation(s)
- Zhanyu Wang
- Department of Surgical Oncology (Breast Center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qianjin Jiang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chenfang Dong
- Department of Surgical Oncology (Breast Center) of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
40
|
Harguindey S, Alfarouk K, Polo Orozco J, Hardonnière K, Stanciu D, Fais S, Devesa J. A New and Integral Approach to the Etiopathogenesis and Treatment of Breast Cancer Based upon Its Hydrogen Ion Dynamics. Int J Mol Sci 2020; 21:E1110. [PMID: 32046158 PMCID: PMC7036897 DOI: 10.3390/ijms21031110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Despite all efforts, the treatment of breast cancer (BC) cannot be considered to be a success story. The advances in surgery, chemotherapy and radiotherapy have not been sufficient at all. Indeed, the accumulated experience clearly indicates that new perspectives and non-main stream approaches are needed to better characterize the etiopathogenesis and treatment of this disease. This contribution deals with how the new pH-centric anticancer paradigm plays a fundamental role in reaching a more integral understanding of the etiology, pathogenesis, and treatment of this multifactorial disease. For the first time, the armamentarium available for the treatment of the different types and phases of BC is approached here from a Unitarian perspective-based upon the hydrogen ion dynamics of cancer. The wide-ranged pH-related molecular, biochemical and metabolic model is able to embrace most of the fields and subfields of breast cancer etiopathogenesis and treatment. This single and integrated approach allows advancing towards a unidirectional, concerted and synergistic program of treatment. Further efforts in this line are likely to first improve the therapeutics of each subtype of this tumor and every individual patient in every phase of the disease.
Collapse
Affiliation(s)
- Salvador Harguindey
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Kévin Hardonnière
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92290 Châtenay-Malabry, France;
| | - Daniel Stanciu
- Scientific Direction, MCS Foundation For Life, 5623KR Eindhoven, The Netherlands;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena, 299, 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, Travesía de Montouto 24, 15886 Teo, Spain;
| |
Collapse
|
41
|
Barbosa AM, Martel F. Targeting Glucose Transporters for Breast Cancer Therapy: The Effect of Natural and Synthetic Compounds. Cancers (Basel) 2020; 12:cancers12010154. [PMID: 31936350 PMCID: PMC7016663 DOI: 10.3390/cancers12010154] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Reprogramming of cellular energy metabolism is widely accepted to be a cancer hallmark. The deviant energetic metabolism of cancer cells-known as the Warburg effect-consists in much higher rates of glucose uptake and glycolytic oxidation coupled with the production of lactic acid, even in the presence of oxygen. Consequently, cancer cells have higher glucose needs and thus display a higher sensitivity to glucose deprivation-induced death than normal cells. So, inhibitors of glucose uptake are potential therapeutic targets in cancer. Breast cancer is the most commonly diagnosed cancer and a leading cause of cancer death in women worldwide. Overexpression of facilitative glucose transporters (GLUT), mainly GLUT1, in breast cancer cells is firmly established, and the consequences of GLUT inhibition and/or knockout are under investigation. Herein we review the compounds, both of natural and synthetic origin, found to interfere with uptake of glucose by breast cancer cells, and the consequences of interference with that mechanism on breast cancer cell biology. We will also present data where the interaction with GLUT is exploited in order to increase the efficiency or selectivity of anticancer agents, in breast cancer cells.
Collapse
Affiliation(s)
- Ana M. Barbosa
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4169-007 Porto, Portugal;
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Correspondence: ; Tel.: +351-22-042-6654
| |
Collapse
|
42
|
Dufresne J, Bowden P, Thavarajah T, Florentinus-Mefailoski A, Chen ZZ, Tucholska M, Norzin T, Ho MT, Phan M, Mohamed N, Ravandi A, Stanton E, Slutsky AS, Dos Santos CC, Romaschin A, Marshall JC, Addison C, Malone S, Heyland D, Scheltens P, Killestein J, Teunissen C, Diamandis EP, Siu KWM, Marshall JG. The plasma peptides of breast versus ovarian cancer. Clin Proteomics 2019; 16:43. [PMID: 31889940 PMCID: PMC6927194 DOI: 10.1186/s12014-019-9262-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background There is a need to demonstrate a proof of principle that proteomics has the capacity to analyze plasma from breast cancer versus other diseases and controls in a multisite clinical trial design. The peptides or proteins that show a high observation frequency, and/or precursor intensity, specific to breast cancer plasma might be discovered by comparison to other diseases and matched controls. The endogenous tryptic peptides of breast cancer plasma were compared to ovarian cancer, female normal, sepsis, heart attack, Alzheimer's and multiple sclerosis along with the institution-matched normal and control samples collected directly onto ice. Methods Endogenous tryptic peptides were extracted from individual breast cancer and control EDTA plasma samples in a step gradient of acetonitrile, and collected over preparative C18 for LC-ESI-MS/MS with a set of LTQ XL linear quadrupole ion traps working together in parallel to randomly and independently sample clinical populations. The MS/MS spectra were fit to fully tryptic peptides or phosphopeptides within proteins using the X!TANDEM algorithm. The protein observation frequency was counted using the SEQUEST algorithm after selecting the single best charge state and peptide sequence for each MS/MS spectra. The observation frequency was subsequently tested by Chi Square analysis. The log10 precursor intensity was compared by ANOVA in the R statistical system. Results Peptides and/or phosphopeptides of common plasma proteins such as APOE, C4A, C4B, C3, APOA1, APOC2, APOC4, ITIH3 and ITIH4 showed increased observation frequency and/or precursor intensity in breast cancer. Many cellular proteins also showed large changes in frequency by Chi Square (χ2 > 100, p < 0.0001) in the breast cancer samples such as CPEB1, LTBP4, HIF-1A, IGHE, RAB44, NEFM, C19orf82, SLC35B1, 1D12A, C8orf34, HIF1A, OCLN, EYA1, HLA-DRB1, LARS, PTPDC1, WWC1, ZNF562, PTMA, MGAT1, NDUFA1, NOGOC, OR1E1, OR1E2, CFI, HSA12, GCSH, ELTD1, TBX15, NR2C2, FLJ00045, PDLIM1, GALNT9, ASH2L, PPFIBP1, LRRC4B, SLCO3A1, BHMT2, CS, FAM188B2, LGALS7, SAT2, SFRS8, SLC22A12, WNT9B, SLC2A4, ZNF101, WT1, CCDC47, ERLIN1, SPFH1, EID2, THOC1, DDX47, MREG, PTPRE, EMILIN1, DKFZp779G1236 and MAP3K8 among others. The protein gene symbols with large Chi Square values were significantly enriched in proteins that showed a complex set of previously established functional and structural relationships by STRING analysis. An increase in mean precursor intensity of peptides was observed for QSER1 as well as SLC35B1, IQCJ-SCHIP1, MREG, BHMT2, LGALS7, THOC1, ANXA4, DHDDS, SAT2, PTMA and FYCO1 among others. In contrast, the QSER1 peptide QPKVKAEPPPK was apparently specific to ovarian cancer. Conclusion There was striking agreement between the breast cancer plasma peptides and proteins discovered by LC-ESI-MS/MS with previous biomarkers from tumors, cells lines or body fluids by genetic or biochemical methods. The results indicate that variation in plasma peptides from breast cancer versus ovarian cancer may be directly discovered by LC-ESI-MS/MS that will be a powerful tool for clinical research. It may be possible to use a battery of sensitive and robust linear quadrupole ion traps for random and independent sampling of plasma from a multisite clinical trial.
Collapse
Affiliation(s)
- Jaimie Dufresne
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Pete Bowden
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Thanusi Thavarajah
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Angelique Florentinus-Mefailoski
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Zhuo Zhen Chen
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Monika Tucholska
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Tenzin Norzin
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Margaret Truc Ho
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Morla Phan
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Nargiz Mohamed
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada
| | - Amir Ravandi
- 2Institute of Cardiovascular Sciences, St. Boniface Hospital Research Center, University of Manitoba, Winnipeg, Canada
| | - Eric Stanton
- 3Division of Cardiology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Arthur S Slutsky
- 4St. Michael's Hospital, Keenan Chair in Medicine, Professor of Medicine, Surgery & Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Claudia C Dos Santos
- 5St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, Canada
| | - Alexander Romaschin
- 5St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, Canada
| | - John C Marshall
- 5St. Michael's Hospital, Keenan Research Centre for Biomedical Science, Toronto, Canada
| | - Christina Addison
- 6Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Shawn Malone
- 6Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Daren Heyland
- 7Clinical Evaluation Research Unit, Kingston General Hospital, Kingston, Canada
| | - Philip Scheltens
- 8Alzheimer Center, Dept of Neurology, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Joep Killestein
- 9MS Center, Dept of Neurology, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Charlotte Teunissen
- 10Neurochemistry Lab and Biobank, Dept of Clinical Chemsitry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | | - K W M Siu
- 12University of Windsor, Windsor, Canada
| | - John G Marshall
- 1Ryerson Analytical Biochemistry Laboratory (RABL), Department of Chemistry and Biology, Faculty of Science, Ryerson University, 350 Victoria St., Toronto, ON Canada.,13International Biobank of Luxembourg (IBBL), Luxembourg Institute of Health (formerly CRP Sante Luxembourg), Strassen, Luxembourg
| |
Collapse
|
43
|
Wu Z, Wu J, Zhao Q, Fu S, Jin J. Emerging roles of aerobic glycolysis in breast cancer. Clin Transl Oncol 2019; 22:631-646. [PMID: 31359335 DOI: 10.1007/s12094-019-02187-8] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022]
Abstract
Altered aerobic glycolysis is a well-recognized characteristic of cancer cell energy metabolism, known as the Warburg effect. Even in the presence of abundant oxygen, a majority of tumor cells produce substantial amounts of energy through a high glycolytic metabolism, and breast cancer (BC) is no exception. Breast cancer continues to be the second leading cause of cancer-associated mortality in women worldwide. However, the precise role of aerobic glycolysis in the development of BC remains elusive. Therefore, the present review attempts to address the implication of key enzymes of the aerobic glycolytic pathway including hexokinase (HK), phosphofructokinase (PFK) and pyruvate kinase (PK), glucose transporters (GLUTs), together with related signaling pathways including protein kinase B(PI3K/AKT), mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) and transcription factors (c-myc, p53 and HIF-1) in the research of BC. Thus, the review of aerobic glycolysis in BC may evoke novel ideas for the BC treatment.
Collapse
Affiliation(s)
- Z Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - J Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Q Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, People's Republic of China
| | - S Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - J Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
44
|
Resveratrol Action on Lipid Metabolism in Cancer. Int J Mol Sci 2019; 20:ijms20112704. [PMID: 31159437 PMCID: PMC6601040 DOI: 10.3390/ijms20112704] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer diseases have the leading position in human mortality nowadays. The age of oncologic patients is still decreasing, and the entire scientific society is eager for new ways to fight against cancer. One of the most discussed issues is prevention by means of natural substances. Resveratrol is a naturally occurring plant polyphenol with proven antioxidant, anti-inflammatory, and anticancer effects. Tumor cells display specific changes in the metabolism of various lipids. Resveratrol alters lipid metabolism in cancer, thereby affecting storage of energy, cell signaling, proliferation, progression, and invasiveness of cancer cells. At the whole organism level, it contributes to the optimal metabolism extent with respect to the demands of the organism. Thus, resveratrol could be used as a preventive and anticancer agent. In this review, we focus on some of the plethora of lipid pathways and signal molecules which are affected by resveratrol during carcinogenesis.
Collapse
|
45
|
Aftab S, Shakoori AR. Low glucose availability alters the expression of genes involved in initial adhesion of human glioblastoma cancer cell line SF767. J Cell Biochem 2019; 120:16824-16839. [PMID: 31111555 DOI: 10.1002/jcb.28940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying the metabolic pathways of cancer cells is considered as a key to control cancer malignancies and open windows for effective drug discovery against cancer. Of all the properties of a tumor, metastasis potential is a defining characteristic. Metastasis is controlled by a variety of factors that directly control the expression of cell adhesion proteins. In this study we have investigated the expression of cell to cell and cell to matrix adhesion protein genes during the initial phases of attachment of human glioblastoma cancer cell line SF767 (66Y old human female: UCSF Neurosurgery Tissue Bank) to the attachment surface under (Cell culture treated polystyrene plate bottom) glucose-rich and glucose-starved conditions. The aim was to imitate the natural microenvironment of glucose availability to cancer cells inside a tumor that triggers epithelial to mesenchymal transition (EMT). In this study, we have observed the gene expression of epithelial and mesenchymal isoforms of cadherin (E-CAD and N-CAD) and Ig like cell adhesion molecules (E-CAM and N-CAM) along with Integrin family subunits for the initial attachment of cancer cells. We observed that high glucose environments promoted cell survival and cell adhesion, whereas low glucose accelerated EMT by downregulating the expression level of integrin, E-CAD, and N-CAD, and upregulation of N-CAM during early period of cell adhesion. Low glucose availability also downregulated variety of structural and regulatory genes, such as zinc finger E-box binding home box 1A), cytokeratin, Snail, and β catenin, and upregulation of hypoxia-inducible factor 1, matrix metalloprotease 13/Collagenase 3, vimentim, p120, and fructose 1,6 bisphosphatase. Glucose conditions are more efficient for cancer studies in this case glioblastoma cells.
Collapse
Affiliation(s)
- Saira Aftab
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore, Pakistan
| | - Abdul Rauf Shakoori
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore, Pakistan.,Department of Biochemistry, Faculty of Life Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
46
|
Dronamraju V, Ibrahim BA, Briski KP, Sylvester PW. γ-Tocotrienol Suppression of the Warburg Effect Is Mediated by AMPK Activation in Human Breast Cancer Cells. Nutr Cancer 2019; 71:1214-1228. [PMID: 30955359 DOI: 10.1080/01635581.2019.1599969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cancer cell metabolism is characterized by aerobic glycolysis or the "Warburg effect". Enhanced Akt signaling is associated with activation of various downstream enzymes involved in the glycolytic process, whereas activation of 5'-AMP-activated kinase (AMPK) acts to terminate energy expending mechanisms and decrease glycolytic enzyme expression. Studies were conducted to determine if the anticancer effects of γ-tocotrienol, are mediated through a suppression in aerobic glycolysis. Results show that treatment with 0-7 μM γ-tocotrienol throughout a 4-day culture period resulted in a dose-responsive increase in AMPK activation, and corresponding decrease in Akt activity in human MCF-7 and MDA-MB-231 breast cancer cells. γ-Tocotrienol treatment was also found to induce a dose-responsive decrease in phosphorylated-Fox03 (inactivated), a transcription factor that acts to inhibit in the levels of glycolytic enzyme, and this decrease was associated with a reduction in glycolytic enzyme levels and activity, as well as glucose consumption in these cells. PCR microarray analysis shows that γ-tocotrienol treatment decreases the expression of genes associate with metabolic signaling and glycolysis in MCF-7 and MDA-MB-231 breast cancer cells. In summary, these findings demonstrate that the anticancer effects of γ-tocotrienol are mediated, at least in part, by a suppression in the Warburg effect.
Collapse
Affiliation(s)
| | - Baher A Ibrahim
- College of Pharmacy, University of Louisiana at Monroe , Monroe , Louisiana , USA
| | - Karen P Briski
- College of Pharmacy, University of Louisiana at Monroe , Monroe , Louisiana , USA
| | - Paul W Sylvester
- College of Pharmacy, University of Louisiana at Monroe , Monroe , Louisiana , USA
| |
Collapse
|
47
|
He Y, Gu Z, Zhu Q, Chen M, He C, Huang Y, Li Q, Di W. CA125 over‐release behavior following a 75‐g oral glucose test as a predictive biomarker of multidrug resistance in patients with ovarian cancer. Int J Cancer 2019; 145:1690-1700. [PMID: 30807642 DOI: 10.1002/ijc.32237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Yifeng He
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Tumor Microenvironment and Metastasis ProgramThe Wistar Institute, University of Pennsylvania Philadelphia PA
| | - Zhuowei Gu
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Qiujing Zhu
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Mo Chen
- Department of GynecologyObstetrics and Gynecology Hospital, Fudan University Shanghai China
| | - Chenghui He
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Yuting Huang
- Children's Research Institute, Children's National Medical Center Washington WA
| | - Qing Li
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- State Key Laboratory of Oncogene and Related GenesShanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| | - Wen Di
- Department of Obstetrics and GynecologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
- State Key Laboratory of Oncogene and Related GenesShanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
48
|
Matsui C, Takatani-Nakase T, Maeda S, Takahashi K. High-Glucose Conditions Promote Anchorage-Independent Colony Growth in Human Breast Cancer MCF-7 Cells. Biol Pharm Bull 2018; 41:1379-1383. [PMID: 30175774 DOI: 10.1248/bpb.b18-00174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that hyperglycemia is connected to the malignant progression of breast cancer; however, the effects of hyperglycemia on tumorigenic potential in breast cancer cells are largely unknown. Here, we demonstrated that the ability of the human breast cancer cell line MCF-7 to undertake anchorage-independent colony growth was significantly enhanced when cultured under high-glucose conditions compared with that under physiological glucose conditions. The high-glucose conditions also promoted phosphorylation of Akt, suggesting that MCF-7 cells cultured in these conditions acquired an increased ability to undergo anchorage-independent growth at least in part through Akt activation, which has been linked to the development of breast cancer. These results raise the possibility that regulation of Akt activity contributes to the tumorigenesis of breast cancer under high-glucose conditions, and we propose that additional analyses of high glucose-induced tumor formation would provide novel strategies for the diagnosis and therapy of breast cancer with hyperglycemia.
Collapse
Affiliation(s)
- Chihiro Matsui
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Tomoka Takatani-Nakase
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Sachie Maeda
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Koichi Takahashi
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| |
Collapse
|
49
|
Efremov YR, Proskurina AS, Potter EA, Dolgova EV, Efremova OV, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Cancer Stem Cells: Emergent Nature of Tumor Emergency. Front Genet 2018; 9:544. [PMID: 30505319 PMCID: PMC6250818 DOI: 10.3389/fgene.2018.00544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
A functional analysis of 167 genes overexpressed in Krebs-2 tumor initiating cells was performed. In the first part of the study, the genes were analyzed for their belonging to one or more of the three groups, which represent the three major phenotypic manifestation of malignancy of cancer cells, namely (1) proliferative self-sufficiency, (2) invasive growth and metastasis, and (3) multiple drug resistance. 96 genes out of 167 were identified as possible contributors to at least one of these fundamental properties. It was also found that substantial part of these genes are also known as genes responsible for formation and/or maintenance of the stemness of normal pluri-/multipotent stem cells. These results suggest that the malignancy is simply the ability to maintain the stem cell specific genes expression profile, and, as a consequence, the stemness itself regardless of the controlling effect of stem niches. In the second part of the study, three stress factors combined into the single concept of "generalized cellular stress," which are assumed to activate the expression of these genes, were defined. In addition, possible mechanisms for such activation were identified. The data obtained suggest the existence of a mechanism for the de novo formation of a pluripotent/stem phenotype in the subpopulation of "committed" tumor cells.
Collapse
Affiliation(s)
- Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana V Efremova
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Oleg S Taranov
- The State Research Center of Virology and Biotechnology Vector, Koltsovo, Russia
| | - Aleksandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
50
|
Urra FA, Muñoz F, Córdova-Delgado M, Ramírez MP, Peña-Ahumada B, Rios M, Cruz P, Ahumada-Castro U, Bustos G, Silva-Pavez E, Pulgar R, Morales D, Varela D, Millas-Vargas JP, Retamal E, Ramírez-Rodríguez O, Pessoa-Mahana H, Pavani M, Ferreira J, Cárdenas C, Araya-Maturana R. FR58P1a; a new uncoupler of OXPHOS that inhibits migration in triple-negative breast cancer cells via Sirt1/AMPK/β1-integrin pathway. Sci Rep 2018; 8:13190. [PMID: 30181620 PMCID: PMC6123471 DOI: 10.1038/s41598-018-31367-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Highly malignant triple-negative breast cancer (TNBC) cells rely mostly on glycolysis to maintain cellular homeostasis; however, mitochondria are still required for migration and metastasis. Taking advantage of the metabolic flexibility of TNBC MDA-MB-231 cells to generate subpopulations with glycolytic or oxidative phenotypes, we screened phenolic compounds containing an ortho-carbonyl group with mitochondrial activity and identified a bromoalkyl-ester of hydroquinone named FR58P1a, as a mitochondrial metabolism-affecting compound that uncouples OXPHOS through a protonophoric mechanism. In contrast to well-known protonophore uncoupler FCCP, FR58P1a does not depolarize the plasma membrane and its effect on the mitochondrial membrane potential and bioenergetics is moderate suggesting a mild uncoupling of OXPHOS. FR58P1a activates AMPK in a Sirt1-dependent fashion. Although the activation of Sirt1/AMPK axis by FR58P1a has a cyto-protective role, selectively inhibits fibronectin-dependent adhesion and migration in TNBC cells but not in non-tumoral MCF10A cells by decreasing β1-integrin at the cell surface. Prolonged exposure to FR58P1a triggers a metabolic reprograming in TNBC cells characterized by down-regulation of OXPHOS-related genes that promote cell survival but comprise their ability to migrate. Taken together, our results show that TNBC cell migration is susceptible to mitochondrial alterations induced by small molecules as FR58P1a, which may have therapeutic implications.
Collapse
Affiliation(s)
- Félix A Urra
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
| | - Felipe Muñoz
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Miguel Córdova-Delgado
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - María Paz Ramírez
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Bárbara Peña-Ahumada
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Melany Rios
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Pablo Cruz
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Rodrigo Pulgar
- Laboratorio de Bioinformática y Expresión Génica, INTA-Universidad de Chile, El Líbano, 5524, Santiago, Chile
| | - Danna Morales
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
| | - Diego Varela
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| | - Juan Pablo Millas-Vargas
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Evelyn Retamal
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Oney Ramírez-Rodríguez
- Campus Río Simpson, University of Aysén, Obispo Vielmo 62, Coyhaique, 5952122, Aysén, Chile
| | - Hernán Pessoa-Mahana
- Departamento de Química Orgánica y Físico-Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Casilla 233, Santiago 1, Chile
| | - Mario Pavani
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Casilla 7, Santiago, Chile
| | - Jorge Ferreira
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Casilla 7, Santiago, Chile
| | - César Cárdenas
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, 93106, United States.
- The Buck Institute for Research on Aging, Novato, CA, 94945, United States.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales and Programa de Investigación Asociativa en Cáncer Gástrico, Universidad de Talca, casilla 747, Talca, Chile.
| |
Collapse
|