1
|
Kansara S, Singh A, Badal AK, Rani R, Baligar P, Garg M, Pandey AK. The emerging regulatory roles of non-coding RNAs associated with glucose metabolism in breast cancer. Semin Cancer Biol 2023; 95:1-12. [PMID: 37364663 DOI: 10.1016/j.semcancer.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 04/20/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Altered energy metabolism is one of the hallmarks of tumorigenesis and essential for fulfilling the high demand for metabolic energy in a tumor through accelerating glycolysis and reprogramming the glycolysis metabolism through the Warburg effect. The dysregulated glucose metabolic pathways are coordinated not only by proteins coding genes but also by non-coding RNAs (ncRNAs) during the initiation and cancer progression. The ncRNAs are responsible for regulating numerous cellular processes under developmental and pathological conditions. Recent studies have shown that various ncRNAs such as microRNAs, circular RNAs, and long noncoding RNAs are extensively involved in rewriting glucose metabolism in human cancers. In this review, we demonstrated the role of ncRNAs in the progression of breast cancer with a focus on outlining the aberrant expression of glucose metabolic pathways. Moreover, we have discussed the existing and probable future applications of ncRNAs to regulate energy pathways along with their importance in the prognosis, diagnosis, and future therapeutics for human breast carcinoma.
Collapse
Affiliation(s)
- Samarth Kansara
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| | - Agrata Singh
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| | - Abhishesh Kumar Badal
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India
| | - Reshma Rani
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, 201313, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida 201313, India
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India; National Institute of Pharmaceutical Education and Research, Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
2
|
Jiang C, Jiang Z, Sha G, Wang D, Tang D. Small extracellular vesicle-mediated metabolic reprogramming: from tumors to pre-metastatic niche formation. Cell Commun Signal 2023; 21:116. [PMID: 37208722 DOI: 10.1186/s12964-023-01136-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
Metastasis, the spread of a tumor or cancer from the primary site of the body to a secondary site, is a multi-step process in cancer progression, accounting for various obstacles in cancer treatment and most cancer-related deaths. Metabolic reprogramming refers to adaptive metabolic changes that occur in cancer cells in the tumor microenvironment (TME) to enhance their survival ability and metastatic potential. Stromal cell metabolism also changes to stimulate tumor proliferation and metastasis. Metabolic adaptations of tumor and non-tumor cells exist not only in the TME but also in the pre-metastatic niche (PMN), a remote TME conducive for tumor metastasis. As a novel mediator in cell-to-cell communication, small extracellular vesicles (sEVs), which have a diameter of 30-150 nm, reprogram metabolism in stromal and cancer cells within the TME by transferring bioactive substances including proteins, mRNAs and miRNAs (microRNAs). sEVs can be delivered from the primary TME to PMN, affecting PMN formation in stroma rewriting, angiogenesis, immunological suppression and matrix cell metabolism by mediating metabolic reprogramming. Herein, we review the functions of sEVs in cancer cells and the TME, how sEVs facilitate PMN establishment to trigger metastasis via metabolic reprogramming, and the prospective applications of sEVs in tumor diagnosis and treatment. Video Abstract.
Collapse
Affiliation(s)
- Chuwen Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu Province, China
| | - Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu Province, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
3
|
Ma S, Chen Y, Quan P, Zhang J, Han S, Wang G, Qi R, Zhang X, Wang F, Yuan J, Yang X, Jia W, Qin W. NPAS2 promotes aerobic glycolysis and tumor growth in prostate cancer through HIF-1A signaling. BMC Cancer 2023; 23:280. [PMID: 36978001 PMCID: PMC10045944 DOI: 10.1186/s12885-023-10685-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/28/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa), one of the common malignant tumors, is the second leading cause of cancer-related deaths in men. The circadian rhythm plays a critical role in disease. Circadian disturbances are often found in patients with tumors and enable to promote tumor development and accelerate its progression. Accumulating evidence suggests that the core clock gene NPAS2 (neuronal PAS domain-containing protein 2) has been implicated in tumors initiation and progression. However, there are few studies on the association between NPAS2 and prostate cancer. The purpose of this paper is to investigate the impact of NPAS2 on cell growth and glucose metabolism in prostate cancer. METHODS Quantitative real-time PCR (qRT-PCR), immunohistochemical (IHC) staining, western blot, GEO (Gene Expression Omnibus) and CCLE (Cancer Cell Line Encyclopedia) databases were used to analyze the expression of NPAS2 in human PCa tissues and various PCa cell lines. Cell proliferation was assessed using MTS, clonogenic assays, apoptotic analyses, and subcutaneous tumor formation experiments in nude mice. Glucose uptake, lactate production, cellular oxygen consumption rate and medium pH were measured to examine the effect of NPAS2 on glucose metabolism. The relation of NPAS2 and glycolytic genes was analyzed based on TCGA (The Cancer Genome Atlas) database. RESULTS Our data showed that NPAS2 expression in prostate cancer patient tissue was elevated compared with that in normal prostate tissue. NPAS2 knockdown inhibited cell proliferation and promoted cell apoptosis in vitro and suppressed tumor growth in a nude mouse model in vivo. NPAS2 knockdown led to glucose uptake and lactate production diminished, oxygen consumption rate and pH elevated. NPAS2 increased HIF-1A (hypoxia-inducible factor-1A) expression, leading to enhanced glycolytic metabolism. There was a positive correlation with the expression of NPAS2 and glycolytic genes, these genes were upregulated with overexpression of NPAS2 while knockdown of NPAS2 led to a lower level. CONCLUSION NPAS2 is upregulated in prostate cancer and promotes cell survival by promoting glycolysis and inhibiting oxidative phosphorylation in PCa cells.
Collapse
Affiliation(s)
- Shuaijun Ma
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Yafan Chen
- Department of Human Movement Science, Xi'an Physical Education University, Xi'an, China
| | - Penghe Quan
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Jingliang Zhang
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Shichao Han
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Guohui Wang
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Ruochen Qi
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Xiaoyan Zhang
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Fuli Wang
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China
| | - Jianlin Yuan
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China.
| | - Xiaojian Yang
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China.
| | - Weijing Jia
- Department of Hematology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China.
| | - Weijun Qin
- Department of Urology, The First Affiliated Hospital of Air Force Medical University, 127 Changle West Road, 710032, Xi 'an, Shaanxi, China.
| |
Collapse
|
4
|
Patil N, Howe O, Cahill P, Byrne HJ. Monitoring and modelling the dynamics of the cellular glycolysis pathway: A review and future perspectives. Mol Metab 2022; 66:101635. [PMID: 36379354 PMCID: PMC9703637 DOI: 10.1016/j.molmet.2022.101635] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The dynamics of the cellular glycolysis pathway underpin cellular function and dysfunction, and therefore ultimately health, disease, diagnostic and therapeutic strategies. Evolving our understanding of this fundamental process and its dynamics remains critical. SCOPE OF REVIEW This paper reviews the medical relevance of glycolytic pathway in depth and explores the current state of the art for monitoring and modelling the dynamics of the process. The future perspectives of label free, vibrational microspectroscopic techniques to overcome the limitations of the current approaches are considered. MAJOR CONCLUSIONS Vibrational microspectroscopic techniques can potentially operate in the niche area of limitations of other omics technologies for non-destructive, real-time, in vivo label-free monitoring of glycolysis dynamics at a cellular and subcellular level.
Collapse
Affiliation(s)
- Nitin Patil
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland; School of Physics and Optometric & Clinical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland.
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland
| |
Collapse
|
5
|
Jin X, Kuang Y, Li L, Li H, Zhao T, He Y, Di C, Kang J, Yuan L, Yu B, Li Q. A positive feedback circuit comprising p21 and HIF-1α aggravates hypoxia-induced radioresistance of glioblastoma by promoting Glut1/LDHA-mediated glycolysis. FASEB J 2022; 36:e22229. [PMID: 35199870 DOI: 10.1096/fj.202101736r] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
The radioresistance induced by hypoxia is the major obstacle in the successful treatment of cancer radiotherapy. p21 was initially identified as a widespread inhibitor of cyclin-dependent kinases, through which mediates the p53-dependent cell cycle G1 phase arrest in response to a variety of stress stimuli. In this study, we discovered a novel function of p21, which participated in the regulation of metabolic pathways under hypoxia. We found that p21 was upregulated in glioblastoma (GBM) cells under hypoxic conditions, which enhanced the radioresistance of GBM cells. In principle, HIF-1α is bound directly to the hypoxia response elements (HREs) of the p21 promoter to enhance its transcription activity, in turn, p21 also promoted the transcription of HIF-1α at the mRNA level and maintained HIF-1α function under oxygen deficiency. The positive correlation between p21 and HIF-1α augmented Glut1/LDHA-mediated glycolysis and aggravated the radioresistance of GBM cells. Thus, our results constructed a positive feedback circuit comprising p21/HIF-1α that might play a key role in enhancing the radioresistance of GBM under hypoxia.
Collapse
Affiliation(s)
- Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Lanhai Nuclear Medicine Research Center, Putian, China
| | - Yanbei Kuang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Linying Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongbin Li
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yufang He
- The Third Hospital of Gansu Province, Lanzhou, China
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Lanhai Nuclear Medicine Research Center, Putian, China
| | - Jian Kang
- College of Energy and Power Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Lingyan Yuan
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Lanhai Nuclear Medicine Research Center, Putian, China
| |
Collapse
|
6
|
Lee SH, Golinska M, Griffiths JR. HIF-1-Independent Mechanisms Regulating Metabolic Adaptation in Hypoxic Cancer Cells. Cells 2021; 10:2371. [PMID: 34572020 PMCID: PMC8472468 DOI: 10.3390/cells10092371] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/22/2022] Open
Abstract
In solid tumours, cancer cells exist within hypoxic microenvironments, and their metabolic adaptation to this hypoxia is driven by HIF-1 transcription factor, which is overexpressed in a broad range of human cancers. HIF inhibitors are under pre-clinical investigation and clinical trials, but there is evidence that hypoxic cancer cells can adapt metabolically to HIF-1 inhibition, which would provide a potential route for drug resistance. Here, we review accumulating evidence of such adaptions in carbohydrate and creatine metabolism and other HIF-1-independent mechanisms that might allow cancers to survive hypoxia despite anti-HIF-1 therapy. These include pathways in glucose, glutamine, and lipid metabolism; epigenetic mechanisms; post-translational protein modifications; spatial reorganization of enzymes; signalling pathways such as Myc, PI3K-Akt, 2-hyxdroxyglutarate and AMP-activated protein kinase (AMPK); and activation of the HIF-2 pathway. All of these should be investigated in future work on hypoxia bypass mechanisms in anti-HIF-1 cancer therapy. In principle, agents targeted toward HIF-1β rather than HIF-1α might be advantageous, as both HIF-1 and HIF-2 require HIF-1β for activation. However, HIF-1β is also the aryl hydrocarbon nuclear transporter (ARNT), which has functions in many tissues, so off-target effects should be expected. In general, cancer therapy by HIF inhibition will need careful attention to potential resistance mechanisms.
Collapse
Affiliation(s)
- Shen-Han Lee
- Department of Otorhinolaryngology, Hospital Sultanah Bahiyah, KM6 Jalan Langgar, Alor Setar 05460, Kedah, Malaysia
| | - Monika Golinska
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; (M.G.); (J.R.G.)
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - John R. Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; (M.G.); (J.R.G.)
| |
Collapse
|
7
|
Sobiepanek A, Paone A, Cutruzzolà F, Kobiela T. Biophysical characterization of melanoma cell phenotype markers during metastatic progression. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:523-542. [PMID: 33730175 PMCID: PMC8190004 DOI: 10.1007/s00249-021-01514-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/30/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Melanoma is the most fatal form of skin cancer, with increasing prevalence worldwide. The most common melanoma genetic driver is mutation of the proto-oncogene serine/threonine kinase BRAF; thus, the inhibition of its MAP kinase pathway by specific inhibitors is a commonly applied therapy. However, many patients are resistant, or develop resistance to this type of monotherapy, and therefore combined therapies which target other signaling pathways through various molecular mechanisms are required. A possible strategy may involve targeting cellular energy metabolism, which has been recognized as crucial for cancer development and progression and which connects through glycolysis to cell surface glycan biosynthetic pathways. Protein glycosylation is a hallmark of more than 50% of the human proteome and it has been recognized that altered glycosylation occurs during the metastatic progression of melanoma cells which, in turn facilitates their migration. This review provides a description of recent advances in the search for factors able to remodel cell metabolism between glycolysis and oxidative phosphorylation, and of changes in specific markers and in the biophysical properties of cells during melanoma development from a nevus to metastasis. This development is accompanied by changes in the expression of surface glycans, with corresponding changes in ligand-receptor affinity, giving rise to structural features and viscoelastic parameters particularly well suited to study by label-free biophysical methods.
Collapse
Affiliation(s)
- Anna Sobiepanek
- Laboratory of Biomolecular Interactions Studies, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland.
| | - Alessio Paone
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Tomasz Kobiela
- Laboratory of Biomolecular Interactions Studies, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| |
Collapse
|
8
|
Li T, Xian HC, Dai L, Tang YL, Liang XH. Tip of the Iceberg: Roles of CircRNAs in Cancer Glycolysis. Onco Targets Ther 2021; 14:2379-2395. [PMID: 33854335 PMCID: PMC8039208 DOI: 10.2147/ott.s297140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/24/2021] [Indexed: 02/05/2023] Open
Abstract
Warburg effect reflects that tumor cells tend to generate energy by aerobic glycolysis rather than oxidative phosphorylation (OXPHOS), thus promoting the development of malignant tumors. As a kind of non-coding RNA, circular RNA (circRNA) is characterized by a closed ring structure and emerges as a regulator of cancer metabolism. Mounting studies revealed that circRNA can regulate the cancer metabolism process through affecting the expression of glycolysis relevant enzymes, transcription factors (TFs), and signaling pathways. In this review, we comprehensively analyzed and concluded the mechanism of circRNA regulating glycolysis, hoping to deepen the cognition of the cancer metabolic regulatory network and to reap huge fruits in targeted cancer treatment.
Collapse
Affiliation(s)
- Tan Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Hong-Chun Xian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Li Dai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
9
|
Yang Y, Liu L, Sun J, Wang S, Yang Z, Li H, Huang N, Zhao W. Deoxypodophyllotoxin Inhibits Non-Small Cell Lung Cancer Cell Growth by Reducing HIF-1α-Mediated Glycolysis. Front Oncol 2021; 11:629543. [PMID: 33732648 PMCID: PMC7959795 DOI: 10.3389/fonc.2021.629543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cell proliferation is a metabolically demanding process that requires high rate of glycolysis to support anabolic growth. Deoxypodophyllotoxin (DPT) is a natural flavonolignan with various pharmacological activities, including antitumor effect. However, whether DPT affects the metabolic reprogramming of cancer cells is unknown. The purpose of this study is to investigate the role of DPT on non-small cell lung cancer (NSCLC) and to explore whether HIF-1α-mediated glycolysis is involved in its mechanism of action.The level of HIF-1α mRNA and protein in NSCLC cells following DPT treatment was detected using qRT-PCR and western blotting, respectively. Cell Counting Kit-8 (CCK-8) and caspase-3 activity assays were performed to analyze cell proliferation and apoptosis. The underlying molecular mechanism was identified by dual luciferase assay, Western blotting, qRT-PCR, glucose consumption, lactate production, and immunoprecipitation. A murine NSCLC model was used to clarify the effect of DPT treatment on tumor cell proliferation. Our findings showed that DPT treatment inhibited NSCLC cell growth in a dose- and time-dependent manner. Further analysis suggested that DPT treatment inhibited HIF-1α signaling pathway by Parkin-mediated protein degradation in NSCLC cells. DPT treatment significantly decreased glucose consumption and lactate production. In addition, DPT treatment reduced the expression of HIF-1α target genes, including GLUT1, HK2 and LDHA, resulting in reduction in glycolysis. We further revealed that DPT-induced cell growth inhibition and increased glucose and lactate levels could be reversed by overexpressing HIF-1α. Additionally, we found that DPT repressed NSCLC growth and GLUT1, HK2 and LDHA expression in vivo. Overall, this study suggested that DPT inhibited NSCLC growth by preventing HIF-1α-mediated glycolysis.
Collapse
Affiliation(s)
- Yuping Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Lingling Liu
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China.,Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Jinghui Sun
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China
| | - Shu Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | | | - Honghui Li
- Department of Refractive Surgery, Chengdu Aier Eye Hospital, Chengdu, China
| | - Na Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wei Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.,School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China
| |
Collapse
|
10
|
Zarisfi M, Nguyen T, Nedrow JR, Le A. The Heterogeneity Metabolism of Renal Cell Carcinomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:117-126. [PMID: 34014538 DOI: 10.1007/978-3-030-65768-0_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
According to data from the American Cancer Society, cancer is one of the deadliest health problems globally. Annually, renal cell carcinoma (RCC) causes more than 100,000 deaths worldwide [1-4], posing an urgent need to develop effective treatments to increase patient survival outcomes. New therapies are expected to address a major factor contributing to cancer's resistance to standard therapies: oncogenic heterogeneity. Gene expression can vary tremendously among different types of cancers, different patients of the same tumor type, and even within individual tumors; various metabolic phenotypes can emerge, making singletherapy approaches insufficient. Novel strategies targeting the diverse metabolism of cancers aim to overcome this obstacle. Though some have yielded positive results, it remains a challenge to uncover all of the distinct metabolic profiles of RCC. In the quest to overcome this obstacle, the metabolic oriented research focusing on these cancers has offered freshly new perspectives, which are expected to contribute heavily to the development of new treatments.
Collapse
Affiliation(s)
- Mohammadreza Zarisfi
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tu Nguyen
- University of California, Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jessie R Nedrow
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Le
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA.
| |
Collapse
|
11
|
Zhang D, Xu X, Ye Q. Metabolism and immunity in breast cancer. Front Med 2020; 15:178-207. [PMID: 33074528 DOI: 10.1007/s11684-020-0793-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022]
Abstract
Breast cancer is one of the most common malignancies that seriously threaten women's health. In the process of the malignant transformation of breast cancer, metabolic reprogramming and immune evasion represent the two main fascinating characteristics of cancer and facilitate cancer cell proliferation. Breast cancer cells generate energy through increased glucose metabolism. Lipid metabolism contributes to biological signal pathways and forms cell membranes except energy generation. Amino acids act as basic protein units and metabolic regulators in supporting cell growth. For tumor-associated immunity, poor immunogenicity and heightened immunosuppression cause breast cancer cells to evade the host's immune system. For the past few years, the complex mechanisms of metabolic reprogramming and immune evasion are deeply investigated, and the genes involved in these processes are used as clinical therapeutic targets for breast cancer. Here, we review the recent findings related to abnormal metabolism and immune characteristics, regulatory mechanisms, their links, and relevant therapeutic strategies.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing, 100850, China.
| |
Collapse
|
12
|
Lee SH, Griffiths JR. How and Why Are Cancers Acidic? Carbonic Anhydrase IX and the Homeostatic Control of Tumour Extracellular pH. Cancers (Basel) 2020; 12:cancers12061616. [PMID: 32570870 PMCID: PMC7352839 DOI: 10.3390/cancers12061616] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
The acidic tumour microenvironment is now recognized as a tumour phenotype that drives cancer somatic evolution and disease progression, causing cancer cells to become more invasive and to metastasise. This property of solid tumours reflects a complex interplay between cellular carbon metabolism and acid removal that is mediated by cell membrane carbonic anhydrases and various transport proteins, interstitial fluid buffering, and abnormal tumour-associated vessels. In the past two decades, a convergence of advances in the experimental and mathematical modelling of human cancers, as well as non-invasive pH-imaging techniques, has yielded new insights into the physiological mechanisms that govern tumour extracellular pH (pHe). In this review, we examine the mechanisms by which solid tumours maintain a low pHe, with a focus on carbonic anhydrase IX (CAIX), a cancer-associated cell surface enzyme. We also review the accumulating evidence that suggest a role for CAIX as a biological pH-stat by which solid tumours stabilize their pHe. Finally, we highlight the prospects for the clinical translation of CAIX-targeted therapies in oncology.
Collapse
Affiliation(s)
- Shen-Han Lee
- Department of Otorhinolaryngology, Hospital Sultanah Bahiyah, Jalan Langgar, Alor Setar 05460, Kedah, Malaysia
- Correspondence:
| | - John R. Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK;
| |
Collapse
|
13
|
Inhibition of Kv10.1 Channels Sensitizes Mitochondria of Cancer Cells to Antimetabolic Agents. Cancers (Basel) 2020; 12:cancers12040920. [PMID: 32283712 PMCID: PMC7226288 DOI: 10.3390/cancers12040920] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Reprogramming of energy metabolism constitutes one of the hallmarks of cancer and is, therefore, an emerging therapeutic target. We describe here that the potassium channel Kv10.1, which is frequently overexpressed in primary and metastatic cancer, and has been proposed a therapeutic target, participates in metabolic adaptation of cancer cells through regulation of mitochondrial dynamics. We used biochemical and cell biological techniques, live cell imaging and high-resolution microscopy, among other approaches, to study the impact of Kv10.1 on the regulation of mitochondrial stability. Inhibition of Kv10.1 expression or function led to mitochondrial fragmentation, increase in reactive oxygen species and increased autophagy. Cells with endogenous overexpression of Kv10.1 were also more sensitive to mitochondrial metabolism inhibitors than cells with low expression, indicating that they are more dependent on mitochondrial function. Consistently, a combined therapy using functional monoclonal antibodies for Kv10.1 and mitochondrial metabolism inhibitors resulted in enhanced efficacy of the inhibitors. Our data reveal a new mechanism regulated by Kv10.1 in cancer and a novel strategy to overcome drug resistance in cancers with a high expression of Kv10.1.
Collapse
|
14
|
Logozzi M, Spugnini E, Mizzoni D, Di Raimo R, Fais S. Extracellular acidity and increased exosome release as key phenotypes of malignant tumors. Cancer Metastasis Rev 2020; 38:93-101. [PMID: 30715644 DOI: 10.1007/s10555-019-09783-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor milieu is characteristically acidic as a consequence of the fermentative metabolism of glucose that results in massive accumulation of lactic acid within the cytoplasm. Tumor cells get rid of excessive protons through exchangers that are responsible for the extracellular acidification that selects cellular clones that are more apt at surviving in this challenging and culling environment. Extracellular vesicles (EVs) are vesicles with diameters ranging from nm to μm that are released from the cells to deliver nucleic acids, proteins, and lipids to adjacent or distant cells. EVs are involved in a plethora of biological events that promote tumor progression including unrestricted proliferation, angiogenesis, migration, local invasion, preparation of the metastatic niche, metastasis, downregulation or hijacking of the immune system, and drug resistance. There is evidence that the release of specific exosomes is increased many folds in cancer patients, as shown by many techniques aimed at evaluating "liquid biopsies". The quality of the exosomal contents has been shown to vary at the different moments of tumor life such as local invasion or metastasis. In vitro studies have recently pointed out that cancer acidity is a major determinant in inducing increased exosome release by human cancer cells, by showing that exosomal release was increased as the pH moved from 7.4 pH to the typical pH of cancer that is 6.5. In this review, we emphasize the recent evidence that tumor acidity and exosomes levels are strictly related and strongly contribute to the malignant tumor phenotypes.
Collapse
Affiliation(s)
- Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Enrico Spugnini
- SAFU Department, Regina Elena Cancer Institute, Via Elio Chianesi 51, 00144, Rome, Italy
| | - Davide Mizzoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Rossella Di Raimo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
15
|
Yuan P, Yang T, Mu J, Zhao J, Yang Y, Yan Z, Hou Y, Chen C, Xing J, Zhang H, Li J. Circadian clock gene NPAS2 promotes reprogramming of glucose metabolism in hepatocellular carcinoma cells. Cancer Lett 2020; 469:498-509. [DOI: 10.1016/j.canlet.2019.11.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
|
16
|
Lee YZ, Guo HC, Zhao GH, Yang CW, Chang HY, Yang RB, Chen L, Lee SJ. Tylophorine-based compounds are therapeutic in rheumatoid arthritis by targeting the caprin-1 ribonucleoprotein complex and inhibiting expression of associated c-Myc and HIF-1α. Pharmacol Res 2019; 152:104581. [PMID: 31794869 DOI: 10.1016/j.phrs.2019.104581] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 12/31/2022]
Abstract
Interruption of the Warburg effect - the observation that un-stimulated macrophages reprogram their core metabolism from oxidative phosphorylation toward aerobic glycolysis to become pro-inflammatory M1 macrophages upon stimulation - is an emerging strategy for the treatment of cancer and anti-inflammatory diseases such as rheumatoid arthritis. We studied this process with view to the discovery of novel therapeutics, and found that tylophorine-based compounds targeted a ribonucleoprotein complex containing caprin-1 and mRNAs of c-Myc and HIF-1α in LPS/IFN-γ stimulated Raw264.7 cells, diminished the protein levels of c-Myc and HIF-1α, and consequently downregulated their targeted genes that are associated with the Warburg effect, as well as the pro-inflammatory iNOS and COX2. The tylophorine-based compound DBQ 33b significantly meliorated the severity and incidence of type II collagen-monoclonal antibody-induced rheumatoid arthritis and diminished gene expressions of c-Myc, HIF-1α, iNOS, COX2, TNFα, and IL-17A in vivo. Moreover, pharmacological inhibition of either c-Myc or HIF-1α exhibited similar effects as the tylophorine-based compound DBQ 33b, even though inhibition of c-Myc reversed the induction of iNOS and COX2 in LPS/IFN-γ stimulated Raw264.7 cells to a lesser degree. Therefore, simultaneous inhibition of both c-Myc and HIF-1α is efficacious for anti-inflammation in vitro and in vivo and merits further study.
Collapse
Affiliation(s)
- Yue-Zhi Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Huan-Chen Guo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Guan-Hao Zhao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC; Department of Life Sciences, National Central University, Taoyuan, 32001, Taiwan, ROC
| | - Cheng-Wei Yang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Hsin-Yu Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan, ROC
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Shiow-Ju Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan, ROC.
| |
Collapse
|
17
|
Yu T, Wang Y, Fan Y, Fang N, Wang T, Xu T, Shu Y. CircRNAs in cancer metabolism: a review. J Hematol Oncol 2019; 12:90. [PMID: 31484561 PMCID: PMC6727394 DOI: 10.1186/s13045-019-0776-8] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Altered energy metabolism is a hallmark of tumors aiming at supplying necessary nutrients for tumorigenesis and development. These redirected metabolic pathways associated with carbohydrate, lipid and amino acid are orchestrated not only by carcinogenic proteins but by non-coding RNAs. Among them, circular RNA (circRNA), as a kind of novel identified non-coding RNAs, has become the focus of attention. Through binding with corresponding microRNAs or directly contacting proteins, circRNA plays a primarily important role in regulating cellular metabolism. Herein, we analyze the emerging findings and select circRNAs contributing to mutant glycolysis, lipogenesis and lipolysis, glutam inolysis, and oxidative respiration to deepen the understanding about the cancer metabolic regulatory network. In addition, we also discuss the possibility of circRNAs exerting their functions via exosomes and cancer stem cells. Owing to their unique structures and wide impacts, circRNAs may help reap huge fruits in developing clinical treatments targeting cancer metabolism.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, China
| | - Yanfen Wang
- Department of Pathology, The Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Yu Fan
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
| | - Na Fang
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, China
| | - Tongshan Wang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, China
| | - Tongpeng Xu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, China.
| | - Yongqian Shu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
18
|
Gao Y, Yang F, Yang X, Zhang L, Yu H, Cheng X, Xu S, Pan J, Wang K, Li P. Mitochondrial metabolism is inhibited by the
HIF
1α‐
MYC
‐
PGC
‐1β axis in
BRAF
V600E thyroid cancer. FEBS J 2019; 286:1420-1436. [DOI: 10.1111/febs.14786] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 11/17/2018] [Accepted: 02/13/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Yanyan Gao
- Key Laboratory of Nuclear Medicine Ministry of Health Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi China
- Center for Vascular Biology Institute for Translational Medicine College of Medicine Qingdao University China
| | - Fang Yang
- Center of System Medicine Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Xiu‐An Yang
- School of Basic Medical Science Chengde Medical University China
| | - Li Zhang
- Key Laboratory of Nuclear Medicine Ministry of Health Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi China
| | - Huixin Yu
- Key Laboratory of Nuclear Medicine Ministry of Health Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi China
| | - Xian Cheng
- Key Laboratory of Nuclear Medicine Ministry of Health Jiangsu Key Laboratory of Molecular Nuclear Medicine Jiangsu Institute of Nuclear Medicine Wuxi China
| | - Shichen Xu
- State Key Laboratory of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi China
| | - Jie Pan
- State Key Laboratory of Food Science and Technology School of Food Science and Technology Jiangnan University Wuxi China
| | - Kun Wang
- Center for Developmental Cardiology Institute for Translational Medicine College of Medicine Qingdao University Qingdao China
| | - Peifeng Li
- Center for Vascular Biology Institute for Translational Medicine College of Medicine Qingdao University China
| |
Collapse
|
19
|
Lee SH, McIntyre D, Honess D, Hulikova A, Pacheco-Torres J, Cerdán S, Swietach P, Harris AL, Griffiths JR. Carbonic anhydrase IX is a pH-stat that sets an acidic tumour extracellular pH in vivo. Br J Cancer 2018; 119:622-630. [PMID: 30206370 PMCID: PMC6162214 DOI: 10.1038/s41416-018-0216-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/26/2018] [Accepted: 07/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumour carbonic anhydrase IX (CAIX), a hypoxia-inducible tumour-associated cell surface enzyme, is thought to acidify the tumour microenvironment by hydrating CO2 to form protons and bicarbonate, but there is no definitive evidence for this in solid tumours in vivo. METHODS We used 1H magnetic resonance spectroscopic imaging (MRSI) of the extracellular pH probe imidazolyl succinic acid (ISUCA) to measure and spatially map extracellular pH in HCT116 tumours transfected to express CAIX and empty vector controls in SCID mice. We also measured intracellular pH in situ with 31P MRS and measured lactate in freeze-clamped tumours. RESULTS CAIX-expressing tumours had 0.15 pH-unit lower median extracellular pH than control tumours (pH 6.71 tumour vs pH 6.86 control, P = 0.01). Importantly, CAIX expression imposed an upper limit for tumour extracellular pH at 6.93. Despite the increased lactate concentration in CAIX-expressing tumours, 31P MRS showed no difference in intracellular pH, suggesting that CAIX acidifies only the tumour extracellular space. CONCLUSIONS CAIX acidifies the tumour microenvironment, and also provides an extracellular pH control mechanism. We propose that CAIX thus acts as an extracellular pH-stat, maintaining an acidic tumour extracellular pH that is tolerated by cancer cells and favours invasion and metastasis.
Collapse
Affiliation(s)
- Shen-Han Lee
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Singapore Health Services (SingHealth), Department of General Surgery, Singapore General Hospital, Academia, College Road, Singapore, 169856, Singapore
| | - Dominick McIntyre
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Davina Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy & Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Jesús Pacheco-Torres
- Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Traylor 211, Baltimore, MD, 21205, USA
| | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas "Alberto Sols", c/ Arturo Duperier 4, Madrid, 28029, Spain
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Adrian L Harris
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
20
|
Nguyen T, Le A. The Metabolism of Renal Cell Carcinomas and Liver Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1063:107-118. [DOI: 10.1007/978-3-319-77736-8_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Proietti S, Cucina A, Minini M, Bizzarri M. Melatonin, mitochondria, and the cancer cell. Cell Mol Life Sci 2017; 74:4015-4025. [PMID: 28785807 PMCID: PMC11107593 DOI: 10.1007/s00018-017-2612-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/03/2017] [Indexed: 12/12/2022]
Abstract
The long-recognized fact that oxidative stress within mitochondria is a hallmark of mitochondrial dysfunction has stimulated the development of mitochondria-targeted antioxidant therapies. Melatonin should be included among the pharmacological agents able to modulate mitochondrial functions in cancer, given that a number of relevant melatonin-dependent effects are triggered by targeting mitochondrial functions. Indeed, melatonin may modulate the mitochondrial respiratory chain, thus antagonizing the cancer highly glycolytic bioenergetic pathway of cancer cells. Modulation of the mitochondrial respiratory chain, together with Ca2+ release and mitochondrial apoptotic effectors, may enhance the spontaneous or drug-induced apoptotic processes. Given that melatonin may efficiently counteract the Warburg effect while stimulating mitochondrial differentiation and mitochondrial-based apoptosis, it is argued that the pineal neurohormone could represent a promising new perspective in cancer treatment strategy.
Collapse
Affiliation(s)
- Sara Proietti
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Alessandra Cucina
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Mirko Minini
- Department of Surgery, "Pietro Valdoni", Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
22
|
Szaryńska M, Olejniczak A, Kobiela J, Spychalski P, Kmieć Z. Therapeutic strategies against cancer stem cells in human colorectal cancer. Oncol Lett 2017; 14:7653-7668. [PMID: 29250169 PMCID: PMC5727596 DOI: 10.3892/ol.2017.7261] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third most frequent malignancy and represents the fourth most common cause of cancer-associated mortalities in the world. Despite many advances in the treatment of CRC, the 5-year survival rate of patients with CRC remains unsatisfactory due to tumor recurrence and metastases. Recently, cancer stem cells (CSCs), have been suggested to be responsible for the initiation and relapse of the disease, and have been identified in CRC. Due to their basic biological features, which include self-renewal and pluripotency, CSCs may be novel therapeutic targets for CRC and other cancer types. Conventional therapeutics only act on proliferating and mature cancer cells, while quiescent CSCs survive and often become resistant to chemotherapy. In this review, markers of CRC-CSCs are evaluated and the recently introduced experimental therapies that specifically target these cells by inducing CSC proliferation, differentiation and sensitization to apoptotic signals via molecules including Dickkopf-1, bone morphogenetic protein 4, Kindlin-1, tankyrases, and p21-activated kinase 1, are discussed. In addition, novel strategies aimed at inhibiting some crucial processes engaged in cancer progression regulated by the Wnt, transforming growth factor β and Notch signaling pathways (pyrvinium pamoate, silibinin, PRI-724, P17, and P144 peptides) are also evaluated. Although the metabolic alterations in cancer were first described decades ago, it is only recently that the concept of targeting key regulatory molecules of cell metabolism, such as sirtuin 1 (miR-34a) and AMPK (metformin), has emerged. In conclusion, the discovery of CSCs has resulted in the definition of novel therapeutic targets and the development of novel experimental therapies for CRC. However, further investigations are required in order to apply these novel drugs in human CRC.
Collapse
Affiliation(s)
- Magdalena Szaryńska
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Agata Olejniczak
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| | - Jarosław Kobiela
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Piotr Spychalski
- Department of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Medical University of Gdańsk, 80-210 Gdańsk; Gdańsk, Poland
| |
Collapse
|
23
|
Chen SW, Lin YC, Chen RY, Hsieh TC, Yen KY, Liang JA, Yang SN, Wang YC, Chen YH, Chow NH, Kao CH. Immunohistochemical overexpression of hypoxia-induced factor 1α associated with slow reduction in 18fluoro-2-deoxy-D-glucose uptake for chemoradiotherapy in patients with pharyngeal cancer. Eur J Nucl Med Mol Imaging 2016; 43:2343-2352. [PMID: 27311919 DOI: 10.1007/s00259-016-3436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND This study examined genomic factors associated with a reduction in 18fluoro-2-deoxy-D-glucose (FDG) uptake during positron emission tomography-computed tomography (PET-CT) for definitive chemoradiotherapy (CRT) in patients with pharyngeal cancer. METHODS The pretreatment and interim PET-CT images of 25 patients with advanced pharyngeal cancers receiving definitive CRT were prospectively evaluated. The maximum standardized uptake value (SUVmax) of the interim PET-CT and the reduction ratio of the SUVmax (SRR) between the two images were measured. Genomic data from pretreatment incisional biopsy specimens (SLC2A1, CAIX, VEGF, HIF1A, BCL2, Claudin-4, YAP1, MET, MKI67, and EGFR) were analyzed using tissue microarrays. Differences in FDG uptake and SRRs between tumors with low and high gene expression were examined using the Mann-Whitney test. Cox regression analysis was performed to examine the effects of variables on local control. RESULTS The SRR of the primary tumors (SRR-P) was 0.59 ± 0.31, whereas the SRR of metastatic lymph nodes (SRR-N) was 0.54 ± 0.32. Overexpression of HIF1A was associated with a high iSUVmax of the primary tumor (P < 0.001) and neck lymph node (P = 0.04) and a low SRR-P (P = 0.02). Multivariate analysis revealed that patients who had tumors with low SRR-P or high HIF1A expression levels showed inferior local control. CONCLUSION In patients with pharyngeal cancer requiring CRT, HIF1A overexpression was positively associated with high interim SUVmax or a slow reduction in FDG uptake. Prospective trials are needed to determine whether the local control rate can be stratified using the HIF1A level as a biomarker and SRR-P.
Collapse
Affiliation(s)
- Shang-Wen Chen
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan.,School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chun Lin
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan.,The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Rui-Yun Chen
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Te-Chun Hsieh
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Kuo-Yang Yen
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 40447, Taiwan
| | - Shih-Neng Yang
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan.,Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Yao-Ching Wang
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Ya-Huey Chen
- Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Nan-Haw Chow
- Department of Pathology, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hung Kao
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan. .,Graduate Institute of Clinical Medical Science, School of Medicine, College of Medicine, China Medical University, No. 2, Yuh-Der Road, Taichung, 40447, Taiwan.
| |
Collapse
|
24
|
de Vallière C, Cosin-Roger J, Simmen S, Atrott K, Melhem H, Zeitz J, Madanchi M, Tcymbarevich I, Fried M, Kullak-Ublick GA, Vavricka SR, Misselwitz B, Seuwen K, Wagner CA, Eloranta JJ, Rogler G, Ruiz PA. Hypoxia Positively Regulates the Expression of pH-Sensing G-Protein-Coupled Receptor OGR1 (GPR68). Cell Mol Gastroenterol Hepatol 2016; 2:796-810. [PMID: 28174749 PMCID: PMC5247318 DOI: 10.1016/j.jcmgh.2016.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/17/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS A novel family of proton-sensing G-protein-coupled receptors, including ovarian cancer G-protein-coupled receptor 1 (OGR1) (GPR68) has been identified to play a role in pH homeostasis. Hypoxia is known to change tissue pH as a result of anaerobic glucose metabolism through the stabilization of hypoxia-inducible factor-1α. We investigated how hypoxia regulates the expression of OGR1 in the intestinal mucosa and associated cells. METHODS OGR1 expression in murine tumors, human colonic tissue, and myeloid cells was determined by quantitative reverse-transcription polymerase chain reaction. The influence of hypoxia on OGR1 expression was studied in monocytes/macrophages and intestinal mucosa of inflammatory bowel disease (IBD) patients. Changes in OGR1 expression in MonoMac6 (MM6) cells under hypoxia were determined upon stimulation with tumor necrosis factor (TNF), in the presence or absence of nuclear factor-κB (NF-κB) inhibitors. To study the molecular mechanisms involved, chromatin immunoprecipitation analysis of the OGR1 promoter was performed. RESULTS OGR1 expression was significantly higher in tumor tissue compared with normal murine colon tissue. Hypoxia positively regulated the expression of OGR1 in MM6 cells, mouse peritoneal macrophages, primary human intestinal macrophages, and colonic tissue from IBD patients. In MM6 cells, hypoxia-enhanced TNF-induced OGR1 expression was reversed by inhibition of NF-κB. In addition to the effect of TNF and hypoxia, OGR1 expression was increased further at low pH. Chromatin immunoprecipitation analysis showed that HIF-1α, but not NF-κB, binds to the promoter of OGR1 under hypoxia. CONCLUSIONS The enhancement of TNF- and hypoxia-induced OGR1 expression under low pH points to a positive feed-forward regulation of OGR1 activity in acidic conditions, and supports a role for OGR1 in the pathogenesis of IBD.
Collapse
Key Words
- AICAR, 5-aminoimidazole-4-carboxamide-1-β-4-ribofuranoside
- CD, Crohn's disease
- ChIP, chromatin immunoprecipitation
- FCS, fetal calf serum
- GPR, G-protein–coupled receptor
- GRP65
- HIF, hypoxia-inducible factor
- HV, healthy volunteer
- IBD, inflammatory bowel disease
- IEC, intestinal epithelial cell
- IFN, interferon
- IL, interleukin
- Inflammation
- Inflammatory Bowel Disease
- MM6, MonoMac 6
- NF-κB, nuclear factor-κB
- OGR1, ovarian cancer G-protein–coupled receptor 1 (GPR68)
- Ovarian Cancer G-Protein–Coupled Receptor
- RT-qPCR, quantitative reverse-transcription polymerase chain reaction
- SPARC, secreted protein acidic and rich in cysteine
- TDAG8
- TDAG8, T-cell death-associated gene 8 (GPR65)
- TNF, tumor necrosis factor
- Th, T-helper
- UC, ulcerative colitis
- WT, wild type
- mRNA, messenger RNA
Collapse
Affiliation(s)
- Cheryl de Vallière
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jesus Cosin-Roger
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland; Department of Pharmacology and Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Simona Simmen
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Kirstin Atrott
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Hassan Melhem
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jonas Zeitz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Mehdi Madanchi
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Irina Tcymbarevich
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Fried
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Stephan R Vavricka
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Benjamin Misselwitz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University Hospital Zurich, Zurich, Switzerland
| | - Jyrki J Eloranta
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Pedro A Ruiz
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Jang Y, Han J, Kim SJ, Kim J, Lee MJ, Jeong S, Ryu MJ, Seo KS, Choi SY, Shong M, Lim K, Heo JY, Kweon GR. Suppression of mitochondrial respiration with auraptene inhibits the progression of renal cell carcinoma: involvement of HIF-1α degradation. Oncotarget 2016; 6:38127-38. [PMID: 26474388 PMCID: PMC4741988 DOI: 10.18632/oncotarget.5511] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/29/2015] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) progression resulting from the uncontrolled migration and enhanced angiogenesis is an obstacle to effective therapeutic intervention. Tumor metabolism has distinctive feature called Warburg effect, which enhances the aerobic glycolysis rapidly supplying the energy for migration of tumor. To manipulate this metabolic change characteristic of aggressive tumors, we utilized the citrus extract, auraptene, known as a mitochondrial inhibitor, testing its anticancer effects against the RCC4 cell line. We found that auraptene impaired RCC4 cell motility through reduction of mitochondrial respiration and glycolytic pathway-related genes. It also strongly disrupted VEGF-induced angiogenesis in vitro and in vivo. Hypoxia-inducible factor 1a (HIF-1a), a key regulator of cancer metabolism, migration and angiogenesis that is stably expressed in RCCs by virtue of a genetic mutation in the von Hippel–Lindau (VHL) tumor-suppressor protein, was impeded by auraptene, which blocked HIF-1a translation initiation without causing cytotoxicity. We suggest that blockade HIF-1a and reforming energy metabolism with auraptene is an effective approach for suspension RCC progression.
Collapse
Affiliation(s)
- Yunseon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jeongsu Han
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jungim Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Soyeon Jeong
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Kang-Sik Seo
- R&D Center, KT&G Life Sciences, Suwon, Republic of Korea, 443-702
| | - Song-Yi Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Kyu Lim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Brain research institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747.,Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea, 301-747
| |
Collapse
|
26
|
The lncRNA MALAT1, acting through HIF-1α stabilization, enhances arsenite-induced glycolysis in human hepatic L-02 cells. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1685-95. [DOI: 10.1016/j.bbadis.2016.06.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 05/27/2016] [Accepted: 06/05/2016] [Indexed: 02/06/2023]
|
27
|
Sanchez-Sanchez AM, Antolin I, Puente-Moncada N, Suarez S, Gomez-Lobo M, Rodriguez C, Martin V. Melatonin Cytotoxicity Is Associated to Warburg Effect Inhibition in Ewing Sarcoma Cells. PLoS One 2015; 10:e0135420. [PMID: 26252771 PMCID: PMC4529102 DOI: 10.1371/journal.pone.0135420] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/21/2015] [Indexed: 12/30/2022] Open
Abstract
Melatonin kills or inhibits the proliferation of different cancer cell types, and this is associated with an increase or a decrease in reactive oxygen species, respectively. Intracellular oxidants originate mainly from oxidative metabolism, and cancer cells frequently show alterations in this metabolic pathway, such as the Warburg effect (aerobic glycolysis). Thus, we hypothesized that melatonin could also regulate differentially oxidative metabolism in cells where it is cytotoxic (Ewing sarcoma cells) and in cells where it inhibits proliferation (chondrosarcoma cells). Ewing sarcoma cells but not chondrosarcoma cells showed a metabolic profile consistent with aerobic glycolysis, i.e. increased glucose uptake, LDH activity, lactate production and HIF-1α activation. Melatonin reversed Ewing sarcoma metabolic profile and this effect was associated with its cytotoxicity. The differential regulation of metabolism by melatonin could explain why the hormone is harmless for a wide spectrum of normal and only a few tumoral cells, while it kills specific tumor cell types.
Collapse
Affiliation(s)
- Ana M. Sanchez-Sanchez
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Isaac Antolin
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Noelia Puente-Moncada
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Santos Suarez
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
| | - Marina Gomez-Lobo
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Carmen Rodriguez
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Vanesa Martin
- Departamento de Morfología y Biología Celular, Facultad de Medicina, c/Julian Claveria, 33006 Oviedo, University of Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- * E-mail:
| |
Collapse
|
28
|
Effect of Dietary ω-3 Polyunsaturated Fatty Acid DHA on Glycolytic Enzymes and Warburg Phenotypes in Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:137097. [PMID: 26339588 PMCID: PMC4538308 DOI: 10.1155/2015/137097] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/15/2015] [Indexed: 12/22/2022]
Abstract
The omega-3 polyunsaturated fatty acids (ω-3 PUFAs) are a class of lipids that has been shown to have beneficial effects on some chronic degenerative diseases such as cardiovascular diseases, rheumatoid arthritis, inflammatory disorders, diabetes, and cancer. Among ω-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA) has received particular attention for its antiproliferative, proapoptotic, antiangiogenetic, anti-invasion, and antimetastatic properties, even though the involved molecular mechanisms are not well understood. Recently, some in vitro studies showed that DHA promotes the inhibition of glycolytic enzymes and the Warburg phenotype. For example, it was shown that in breast cancer cell lines the modulation of bioenergetic functions is due to the capacity of DHA to activate the AMPK signalling and negatively regulate the HIF-1α functions. Taking into account these considerations, this review is focused on current knowledge concerning the role of DHA in interfering with cancer cell metabolism; this could be considered a further mechanism by which DHA inhibits cancer cell survival and progression.
Collapse
|
29
|
Cabanillas Stanchi KM, Bruchelt G, Handgretinger R, Holzer U. Nifurtimox reduces N-Myc expression and aerobic glycolysis in neuroblastoma. Cancer Biol Ther 2015; 16:1353-63. [PMID: 26177922 DOI: 10.1080/15384047.2015.1070987] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Neuroblastoma is one of the most common solid tumors in childhood and usually accompanied with poor prognosis and rapid tumor progression when diagnosed with amplification of the proto-oncogene N-Myc. The amplification of N-Myc has major influence on the maintenance of aerobic glycolysis, also known as the Warburg effect. This specific switch in the conversion of pyruvate to lactate instead of the conversion of pyruvate to acetyl-coenzyme A even in the presence of oxygen has important benefits for the tumor, e.g. increased production of enzymes and enzyme substrates that are involved in tumor progression, angiogenesis and inhibition of apoptosis. The antiprotozoal drug nifurtimox, which is generally used for the treatment of infections with the parasitic protozoan Trypanosoma cruzi, has been reported to have cytotoxic properties in the therapy of neuroblastoma. However, its action of mechanism has not been described in detail yet. The presented in vitro study on the neuroblastoma cell lines LA-N-1, IMR-32, LS and SK-N-SH shows an increased production of oxidative stress, a reduced lactate dehydrogenase enzyme activity and reduced lactate production after nifurtimox treatment. Furthermore, nifurtimox leads to reduced mRNA and protein levels of the proto-oncogene protein N-Myc. Thus, the current work gives new insights into the effect of nifurtimox on tumor metabolism revealing a shifted glucose metabolism from production of lactate to oxidative phosphorylation and a reduced expression of the major molecular prognostic factor in neuroblastoma N-Myc, presenting nifurtimox as a possible adjuvant therapeutic agent against (high risk) neuroblastoma.
Collapse
Affiliation(s)
- Karin Melanie Cabanillas Stanchi
- a University Hospital Tuebingen; Children's Hospital; Department I - General Pediatrics; Hematology/Oncology ; Tuebingen , Germany
| | - Gernot Bruchelt
- a University Hospital Tuebingen; Children's Hospital; Department I - General Pediatrics; Hematology/Oncology ; Tuebingen , Germany
| | - Rupert Handgretinger
- a University Hospital Tuebingen; Children's Hospital; Department I - General Pediatrics; Hematology/Oncology ; Tuebingen , Germany
| | - Ursula Holzer
- a University Hospital Tuebingen; Children's Hospital; Department I - General Pediatrics; Hematology/Oncology ; Tuebingen , Germany
| |
Collapse
|
30
|
Lemaire L, Franconi F, Siegler B, Legendre C, Garcion E. In vitro expansion of U87-MG human glioblastoma cells under hypoxic conditions affects glucose metabolism and subsequent in vivo growth. Tumour Biol 2015; 36:7699-710. [PMID: 25934335 DOI: 10.1007/s13277-015-3458-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/13/2015] [Indexed: 12/22/2022] Open
Abstract
Hypoxia is a characteristic feature of solid tumors leading to the over expression of hypoxia-inducible factor (HIF)-1α protein and therefore to a specific cellular behavior. However, even though the oxygen tension in tumors is low (<5 %), most of the cell lines used in cancer studies are grown under 21 % oxygen tension. This work focuses on the impact of oxygen conditions during in vitro cell culture on glucose metabolism using 1-(13)C-glucose. Growing U87-MG glioma cells under hypoxic conditions leads to a two- to threefold reduction of labeled glutamine and an accumulation of fructose. However, under both hypoxic and normoxic conditions, glucose is used for de novo synthesis of pyrimidine since the (13)C label is found both in the uracil and ribose moieties. Labeling of the ribose ring demonstrates that U87-MG glioma cells use the reversible branch of the non-oxidative pentose phosphate pathway. Interestingly, stereotactic implantation of U87-MG cells grown under normoxia or mild hypoxia within the striatum of nude mice led to differential growth; the cells grown under hypoxia retaining an imprint of the oxygen adaptation as their development is then slowed down.
Collapse
Affiliation(s)
- L Lemaire
- INSERM U 1066, 'Micro et Nanomédecines Biomimétiques - MINT' IBS - CHU, 4, rue Larrey, 49933, Angers, France. .,LUNAM Université, Université Angers, UMR-S1066, Angers, France.
| | - F Franconi
- PRIMEX, Université d'Angers, LUNAM Université, Angers, France.,PIAM, Université d'Angers, LUNAM Université, Angers, France
| | - B Siegler
- PIAM, Université d'Angers, LUNAM Université, Angers, France
| | - C Legendre
- INSERM U 1066, 'Micro et Nanomédecines Biomimétiques - MINT' IBS - CHU, 4, rue Larrey, 49933, Angers, France.,LUNAM Université, Université Angers, UMR-S1066, Angers, France
| | - E Garcion
- INSERM U 1066, 'Micro et Nanomédecines Biomimétiques - MINT' IBS - CHU, 4, rue Larrey, 49933, Angers, France.,LUNAM Université, Université Angers, UMR-S1066, Angers, France
| |
Collapse
|
31
|
Bluff JE, Reynolds S, Metcalf S, Alizadeh T, Kazan SM, Bucur A, Wholey EG, Bibby BAS, Williams L, Paley MN, Tozer GM. Measurement of the acute metabolic response to hypoxia in rat tumours in vivo using magnetic resonance spectroscopy and hyperpolarised pyruvate. Radiother Oncol 2015; 116:392-9. [PMID: 25824978 PMCID: PMC4612449 DOI: 10.1016/j.radonc.2015.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 03/03/2015] [Accepted: 03/08/2015] [Indexed: 12/11/2022]
Abstract
Purpose To estimate the rate constant for pyruvate to lactate conversion in tumours in response to a hypoxic challenge, using hyperpolarised 13C1-pyruvate and magnetic resonance spectroscopy. Methods and materials Hypoxic inspired gas was used to manipulate rat P22 fibrosarcoma oxygen tension (pO2), confirmed by luminescence decay of oxygen-sensitive probes. Hyperpolarised 13C1-pyruvate was injected into the femoral vein of anaesthetised rats and slice-localised 13C magnetic resonance (MR) spectra acquired. Spectral integral versus time curves for pyruvate and lactate were fitted to a precursor-product model to estimate the rate constant for tumour conversion of pyruvate to lactate (kpl). Mean arterial blood pressure (MABP) and oxygen tension (ArtpO2) were monitored. Pyruvate and lactate concentrations were measured in freeze-clamped tumours. Results MABP, ArtpO2 and tumour pO2 decreased significantly during hypoxia. kpl increased significantly (p < 0.01) from 0.029 ± 0.002 s−1 to 0.049 ± 0.006 s−1 (mean ± SEM) when animals breathing air were switched to hypoxic conditions, whereas pyruvate and lactate concentrations were minimally affected by hypoxia. Both ArtpO2 and MABP influenced the estimate of kpl, with a strong negative correlation between kpl and the product of ArtpO2 and MABP under hypoxia. Conclusion The rate constant for pyruvate to lactate conversion, kpl, responds significantly to a rapid reduction in tumour oxygenation.
Collapse
Affiliation(s)
- Joanne E Bluff
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Steven Reynolds
- Academic Unit of Radiology, Department of Cardiovascular Science, University of Sheffield, UK.
| | - Stephen Metcalf
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Tooba Alizadeh
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Samira M Kazan
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Adriana Bucur
- Academic Unit of Radiology, Department of Cardiovascular Science, University of Sheffield, UK
| | - Emily G Wholey
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Becky A S Bibby
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Leigh Williams
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| | - Martyn N Paley
- Academic Unit of Radiology, Department of Cardiovascular Science, University of Sheffield, UK
| | - Gillian M Tozer
- Tumour Microcirculation Group, Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, UK
| |
Collapse
|
32
|
Analysis and interpretation of transcriptomic data obtained from extended Warburg effect genes in patients with clear cell renal cell carcinoma. Oncoscience 2015; 2:151-86. [PMID: 25859558 PMCID: PMC4381708 DOI: 10.18632/oncoscience.128] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/17/2015] [Indexed: 12/22/2022] Open
Abstract
Background Many cancers adopt a metabolism that is characterized by the well-known Warburg effect (aerobic glycolysis). Recently, numerous attempts have been made to treat cancer by targeting one or more gene products involved in this pathway without notable success. This work outlines a transcriptomic approach to identify genes that are highly perturbed in clear cell renal cell carcinoma (CCRCC). Methods We developed a model of the extended Warburg effect and outlined the model using Cytoscape. Following this, gene expression fold changes (FCs) for tumor and adjacent normal tissue from patients with CCRCC (GSE6344) were mapped on to the network. Gene expression values with FCs of greater than two were considered as potential targets for treatment of CCRCC. Results The Cytoscape network includes glycolysis, gluconeogenesis, the pentose phosphate pathway (PPP), the TCA cycle, the serine/glycine pathway, and partial glutaminolysis and fatty acid synthesis pathways. Gene expression FCs for nine of the 10 CCRCC patients in the GSE6344 data set were consistent with a shift to aerobic glycolysis. Genes involved in glycolysis and the synthesis and transport of lactate were over-expressed, as was the gene that codes for the kinase that inhibits the conversion of pyruvate to acetyl-CoA. Interestingly, genes that code for unique proteins involved in gluconeogenesis were strongly under-expressed as was also the case for the serine/glycine pathway. These latter two results suggest that the role attributed to the M2 isoform of pyruvate kinase (PKM2), frequently the principal isoform of PK present in cancer: i.e. causing a buildup of glucose metabolites that are shunted into branch pathways for synthesis of key biomolecules, may not be operative in CCRCC. The fact that there was no increase in the expression FC of any gene in the PPP is consistent with this hypothesis. Literature protein data generally support the transcriptomic findings. Conclusions A number of key genes have been identified that could serve as valid targets for anti-cancer pharmaceutical agents. Genes that are highly over-expressed include ENO2, HK2, PFKP, SLC2A3, PDK1, and SLC16A1. Genes that are highly under-expressed include ALDOB, PKLR, PFKFB2, G6PC, PCK1, FBP1, PC, and SUCLG1.
Collapse
|
33
|
Choi YB, Harhaj EW. Functional implications of mitochondrial reactive oxygen species generated by oncogenic viruses. ACTA ACUST UNITED AC 2014; 9:423-436. [PMID: 25580106 DOI: 10.1007/s11515-014-1332-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Between 15-20% of human cancers are associated with infection by oncogenic viruses. Oncogenic viruses, including HPV, HBV, HCV and HTLV-1, target mitochondria to influence cell proliferation and survival. Oncogenic viral gene products also trigger the production of reactive oxygen species which can elicit oxidative DNA damage and potentiate oncogenic host signaling pathways. Viral oncogenes may also subvert mitochondria quality control mechanisms such as mitophagy and metabolic adaptation pathways to promote virus replication. Here, we will review recent progress on viral regulation of mitophagy and metabolic adaptation and their roles in viral oncogenesis.
Collapse
Affiliation(s)
- Young Bong Choi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns HopkinsSchool of Medicine, Baltimore, MD 21287, USA
| | - Edward William Harhaj
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns HopkinsSchool of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
34
|
Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, Zhang Q, Wakelam MJO, Karpe F, Schulze A, Harris AL. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep 2014; 9:349-365. [PMID: 25263561 DOI: 10.1016/j.celrep.2014.08.056] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/16/2014] [Accepted: 08/22/2014] [Indexed: 01/22/2023] Open
Abstract
An in vivo model of antiangiogenic therapy allowed us to identify genes upregulated by bevacizumab treatment, including Fatty Acid Binding Protein 3 (FABP3) and FABP7, both of which are involved in fatty acid uptake. In vitro, both were induced by hypoxia in a hypoxia-inducible factor-1α (HIF-1α)-dependent manner. There was a significant lipid droplet (LD) accumulation in hypoxia that was time and O2 concentration dependent. Knockdown of endogenous expression of FABP3, FABP7, or Adipophilin (an essential LD structural component) significantly impaired LD formation under hypoxia. We showed that LD accumulation is due to FABP3/7-dependent fatty acid uptake while de novo fatty acid synthesis is repressed in hypoxia. We also showed that ATP production occurs via β-oxidation or glycogen degradation in a cell-type-dependent manner in hypoxia-reoxygenation. Finally, inhibition of lipid storage reduced protection against reactive oxygen species toxicity, decreased the survival of cells subjected to hypoxia-reoxygenation in vitro, and strongly impaired tumorigenesis in vivo.
Collapse
Affiliation(s)
- Karim Bensaad
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK.
| | - Elena Favaro
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Caroline A Lewis
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Barrie Peck
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Simon Lord
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Jennifer M Collins
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Simon Wigfield
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Francesca M Buffa
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Ji-Liang Li
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Qifeng Zhang
- The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK; NIHR Oxford Biomedical Research Centre, OUH Trust, Churchill Hospital, Oxford OX3 7LF, UK
| | - Almut Schulze
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK; Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany
| | - Adrian L Harris
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| |
Collapse
|
35
|
McCarty MF, Contreras F. Increasing Superoxide Production and the Labile Iron Pool in Tumor Cells may Sensitize Them to Extracellular Ascorbate. Front Oncol 2014; 4:249. [PMID: 25279352 PMCID: PMC4165285 DOI: 10.3389/fonc.2014.00249] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/01/2014] [Indexed: 12/23/2022] Open
Abstract
Low millimolar concentrations of ascorbate are capable of inflicting lethal damage on a high proportion of cancer cells lines, yet leave non-transformed cell lines unscathed. Extracellular generation of hydrogen peroxide, reflecting reduction of molecular oxygen by ascorbate, has been shown to mediate this effect. Although some cancer cell lines express low catalase activity, this cannot fully explain the selective sensitivity of cancer cells to hydrogen peroxide. Ranzato and colleagues have presented evidence for a plausible new explanation of this sensitivity - a high proportion of cancers, via NADPH oxidase complexes or dysfunctional mitochondria, produce elevated amounts of superoxide. This superoxide, via a transition metal-catalyzed transfer of an electron to the hydrogen peroxide produced by ascorbate, can generate deadly hydroxyl radical (Haber-Weiss reaction). It thus can be predicted that concurrent measures which somewhat selectively boost superoxide production in cancers will enhance their sensitivity to i.v. ascorbate therapy. One way to achieve this is to increase the provision of substrate to cancer mitochondria. Measures which inhibit the constitutive hypoxia-inducible factor-1 (HIF-1) activity in cancers (such as salsalate and mTORC1 inhibitors, or an improvement of tumor oxygenation), or that inhibit the HIF-1-inducible pyruvate dehydrogenase kinase (such as dichloroacetate), can be expected to increase pyruvate oxidation. A ketogenic diet should provide more lipid substrate for tumor mitochondria. The cancer-killing activity of 42°C hyperthermia is to some degree contingent on an increase in oxidative stress, likely of mitochondrial origin; reports that hydrogen peroxide synergizes with hyperthermia in killing cancer cells suggest that hyperthermia and i.v. ascorbate could potentiate each other's efficacy. A concurrent enhancement of tumor oxygenation might improve results by decreasing HIF-1 activity while increasing the interaction of ascorbic acid with oxygen. An increased pool of labile iron in cancer cells may contribute to the selective susceptibility of many cancers to i.v. ascorbate; antagonism of NF-kappaB activity with salicylate, and intravenous iron administration, could be employed to further elevate free iron in cancers.
Collapse
|
36
|
Tumor initiating cells and chemoresistance: which is the best strategy to target colon cancer stem cells? BIOMED RESEARCH INTERNATIONAL 2014; 2014:859871. [PMID: 24527460 PMCID: PMC3914574 DOI: 10.1155/2014/859871] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/24/2013] [Indexed: 12/12/2022]
Abstract
There is an emerging body of evidence that chemoresistance and minimal residual disease result from selective resistance of a cell subpopulation from the original tumor that is molecularly and phenotypically distinct. These cells are called “cancer stem cells” (CSCs). In this review, we analyze the potential targeting strategies for eradicating CSCs specifically in order to develop more effective therapeutic strategies for metastatic colon cancer. These include induction of terminal epithelial differentiation of CSCs or targeting some genes expressed only in CSCs and involved in self-renewal and chemoresistance. Ideal targets could be cell regulators that simultaneously control the stemness and the resistance of CSCs. Another important aspect of cancer biology, which can also be harnessed to create novel broad-spectrum anticancer agents, is the Warburg effect, also known as aerobic glycolysis. Actually, little is yet known with regard to the metabolism of CSCs population, leaving an exciting unstudied avenue in the dawn of the emerging field of metabolomics.
Collapse
|
37
|
Yang F, Zhang H, Mei Y, Wu M. Reciprocal regulation of HIF-1α and lincRNA-p21 modulates the Warburg effect. Mol Cell 2013; 53:88-100. [PMID: 24316222 DOI: 10.1016/j.molcel.2013.11.004] [Citation(s) in RCA: 415] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/27/2013] [Accepted: 10/31/2013] [Indexed: 01/06/2023]
Abstract
Hypoxia has long been linked to the Warburg effect, yet the underlying mechanism remains largely unclear. It is also not known if lncRNAs are involved in the contribution of hypoxia to the Warburg effect. Here we show that lincRNA-p21 is a hypoxia-responsive lncRNA and is essential for hypoxia-enhanced glycolysis. Hypoxia/HIF-1α-induced lincRNA-p21 is able to bind HIF-1α and VHL and thus disrupts the VHL-HIF-1α interaction. This disassociation attenuates VHL-mediated HIF-1α ubiquitination and causes HIF-1α accumulation. These data indicate the existence of a positive feedback loop between HIF-1α and lincRNA-p21 that promotes glycolysis under hypoxia. The ability of lincRNA-p21 to promote tumor growth is validated in mouse xenograft models. Together, these findings suggest that lincRNA-p21 is an important player in the regulation of the Warburg effect and also implicate lincRNA-p21 as a valuable therapeutic target for cancer.
Collapse
Affiliation(s)
- Fan Yang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yide Mei
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Mian Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
38
|
Clémençon B, Babot M, Trézéguet V. The mitochondrial ADP/ATP carrier (SLC25 family): pathological implications of its dysfunction. Mol Aspects Med 2013; 34:485-93. [PMID: 23506884 DOI: 10.1016/j.mam.2012.05.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 02/10/2012] [Indexed: 02/04/2023]
Abstract
In aerobic eukaryotic cells, the high energy metabolite ATP is generated mainly within the mitochondria following the process of oxidative phosphorylation. The mitochondrial ATP is exported to the cytoplasm using a specialized transport protein, the ADP/ATP carrier, to provide energy to the cell. Any deficiency or dysfunction of this membrane protein leads to serious consequences on cell metabolism and can cause various diseases such as muscular dystrophy. Described as a decisive player in the programmed cell death, it was recently shown to play a role in cancer. The objective of this review is to summarize the current knowledge of the involvement of the ADP/ATP carrier, encoded by the SLC25A4, SLC25A5, SLC25A6 and SLC25A31 genes, in human diseases and of the efforts made at designing different model systems to study this carrier and the associated pathologies through biochemical, genetic, and structural approaches.
Collapse
Affiliation(s)
- Benjamin Clémençon
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland.
| | | | | |
Collapse
|
39
|
Puszyk WM, Le Trinh T, Chapple S, Liu C. Linking metabolism and epigenetic regulation in development of hepatocellular carcinoma. J Transl Med 2013; 93:983-90. [PMID: 23917878 PMCID: PMC4028619 DOI: 10.1038/labinvest.2013.94] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/07/2013] [Indexed: 12/29/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common form of cancer globally and is rarely curable once detected. The 5-year survival rate of patients diagnosed with late-stage HCC may be as low as 27%. HCC is a cancer largely driven by epigenetic changes that arise from exposure to exogenous environmental factors rather than coding sequence mutations. The liver is susceptible to effects from Hepatitis C and Hepatitis B viruses, exposure to aflatoxin and continuous excessive consumption of alcohol. The liver is a highly metabolic organ balancing many vital biochemical processes; exposure to any of the above environmental factors is associated with loss of liver function and is a major risk factor for the development of HCC. Emerging studies aim to examine the underlying metabolic processes that are abrogated in cancer and lead to the altered flux and availability of key metabolites important for epigenetic processes. Metabolites have been shown to act as substrates for many canonical epigenetic regulators. These enzymes are responsible for regulating histone modification, DNA methylation and micro RNA expression. By studying the impact of altered liver metabolism, we may better understand the long-term epigenetic processes, which lead to the development and progression of HCC.
Collapse
Affiliation(s)
- William Matthew Puszyk
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Thu Le Trinh
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Sarah Chapple
- Cardiovascular Division, BHF Centre of Research Excellence, School of Medicine, King’s College London, London, SE1 9NH, UK
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, USA,Shands Cancer Center, University of Florida, Gainesville, Florida 32610, USA,Correspondence: Chen Liu, M.D., Ph.D., Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, 1600 SW Archer Road, M651, PO 100275, Gainesville, FL 32610. Tel: 352-273-5413; Fax: 352-392-6249
| |
Collapse
|
40
|
Two p53-related metabolic regulators, TIGAR and SCO2, contribute to oroxylin A-mediated glucose metabolism in human hepatoma HepG2 cells. Int J Biochem Cell Biol 2013; 45:1468-78. [DOI: 10.1016/j.biocel.2013.04.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/31/2013] [Accepted: 04/15/2013] [Indexed: 11/15/2022]
|
41
|
Smith TAD, Zanda M, Fleming IN. Hypoxia stimulates 18F-fluorodeoxyglucose uptake in breast cancer cells via hypoxia inducible factor-1 and AMP-activated protein kinase. Nucl Med Biol 2013; 40:858-64. [PMID: 23786679 DOI: 10.1016/j.nucmedbio.2013.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/10/2013] [Accepted: 05/12/2013] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Hypoxia can stimulate (18)F-fluorodeoxyglucose (FDG) uptake in cultured cells. A better understanding of the underlying molecular mechanism is required to determine the value of FDG for studying tumour hypoxia. METHODS The effect of hypoxia on FDG uptake, and key proteins involved in glucose transport and glycolysis, was studied in MCF7 and MDA231 breast cancer cell lines. RESULTS Hypoxia induced a dose- and time-dependent increase in FDG uptake. The FDG increase was transient, suggesting that FDG uptake is only likely to be increased by acute hypoxia (<24 h). Molecular analysis indicated that hypoxia upregulated glut1 and 6-phosphofructo-2-kinase, key proteins involved in regulating glucose transport and glycolysis, and that these changes were induced by Hypoxia-Inducible factor 1 (HIF1) upregulation and/or AMP-activated protein kinase activation. CONCLUSIONS FDG may provide useful information about the oxygenation status of cells in hypoxic regions where HIF1 upregulation is hypoxia-driven.
Collapse
Affiliation(s)
- Tim A D Smith
- Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD
| | | | | |
Collapse
|
42
|
Puglisi MA, Tesori V, Lattanzi W, Gasbarrini GB, Gasbarrini A. Colon cancer stem cells: Controversies and perspectives. World J Gastroenterol 2013; 19:2997-3006. [PMID: 23716979 PMCID: PMC3662939 DOI: 10.3748/wjg.v19.i20.2997] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/25/2013] [Accepted: 04/04/2013] [Indexed: 02/06/2023] Open
Abstract
Tumors have long been viewed as a population in which all cells have the equal propensity to form new tumors, the so called conventional stochastic model. The cutting-edge theory on tumor origin and progression, tends to consider cancer as a stem cell disease. Stem cells are actively involved in the onset and maintenance of colon cancer. This review is intended to examine the state of the art on colon cancer stem cells (CSCs), with regard to the recent achievements of basic research and to the corresponding translational consequences. Specific prominence is given to the hypothesized origin of CSCs and to the methods for their identification. The growing understanding of CSC biology is driving the optimization of novel anti-cancer targeted drugs.
Collapse
|
43
|
Holley AK, Dhar SK, St Clair DK. Curbing cancer's sweet tooth: is there a role for MnSOD in regulation of the Warburg effect? Mitochondrion 2013; 13:170-88. [PMID: 22820117 PMCID: PMC4604438 DOI: 10.1016/j.mito.2012.07.104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 07/04/2012] [Accepted: 07/10/2012] [Indexed: 01/27/2023]
Abstract
Reactive oxygen species (ROS), while vital for normal cellular function, can have harmful effects on cells, leading to the development of diseases such as cancer. The Warburg effect, the shift from oxidative phosphorylation to glycolysis, even in the presence of adequate oxygen, is an important metabolic change that confers many growth and survival advantages to cancer cells. Reactive oxygen species are important regulators of the Warburg effect. The mitochondria-localized antioxidant enzyme manganese superoxide dismutase (MnSOD) is vital to survival in our oxygen-rich atmosphere because it scavenges mitochondrial ROS. MnSOD is important in cancer development and progression. However, the significance of MnSOD in the regulation of the Warburg effect is just now being revealed, and it may significantly impact the treatment of cancer in the future.
Collapse
Affiliation(s)
- Aaron K. Holley
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536
| | - Sanjit Kumar Dhar
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536
| | - Daret K. St Clair
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
44
|
Taddei ML, Giannoni E, Comito G, Chiarugi P. Microenvironment and tumor cell plasticity: an easy way out. Cancer Lett 2013; 341:80-96. [PMID: 23376253 DOI: 10.1016/j.canlet.2013.01.042] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 01/23/2013] [Accepted: 01/24/2013] [Indexed: 12/12/2022]
Abstract
Cancer cells undergo genetic changes allowing their adaptation to environmental changes, thereby obtaining an advantage during the long metastatic route, disseminated of several changes in the surrounding environment. In particular, plasticity in cell motility, mainly due to epigenetic regulation of cancer cells by environmental insults, engage adaptive strategies aimed essentially to survive in hostile milieu, thereby escaping adverse sites. This review is focused on tumor microenvironment as a collection of structural and cellular elements promoting plasticity and adaptive programs. We analyze the role of extracellular matrix stiffness, hypoxia, nutrient deprivation, acidity, as well as different cell populations of tumor microenvironment.
Collapse
Affiliation(s)
- Maria Letizia Taddei
- Department of Biochemical Sciences, University of Florence, Viale Morgagni 50, 50134 Firenze, Italy
| | | | | | | |
Collapse
|
45
|
Pedersen SF, Graebe M, Hag AMF, Hoejgaard L, Sillesen H, Kjaer A. Microvessel density but not neoangiogenesis is associated with 18F-FDG uptake in human atherosclerotic carotid plaques. Mol Imaging Biol 2012; 14:384-92. [PMID: 21732164 DOI: 10.1007/s11307-011-0507-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The vulnerable atherosclerotic lesion exhibits the proliferation of neovessels and inflammation. The imaging modality 2-deoxy-2-[(18)F]fluoro-D: -glucose positron emission tomography ((18)FDG-PET) is considered for the identification of vulnerable plaques. PURPOSE The purpose of this study was to compare the gene expression of neoangiogenesis and vulnerability-associated genes with (18)FDG uptake in patients undergoing carotid endarterectomy. PROCEDURES Human atherosclerotic carotid artery plaques from symptomatic patients were used for gene expression analysis by quantitative PCR of vascular endothelial growth factor (VEGF) and integrin α(V) and integrin β(3) subunits, genes essential during neoangiogenesis. We also evaluated the gene expression of CD34, a measure of microvessel density (MVD), as well as CD68, MMP-9, and cathepsin K, genes of major importance in plaque vulnerability. Gene expression analysis was compared with (18)FDG-PET. RESULTS VEGF and integrin α(V)β(3) gene expression did not correlate with (18)FDG uptake, whereas CD34 gene expression exhibited an inverse correlation with (18)FDG uptake. Additionally, we established that markers of vulnerability were correlated with (18)FDG uptake. CONCLUSIONS Neoangiogenesis is not associated with (18)FDG uptake, whereas MVD and markers of vulnerability correlate with (18)FDG uptake. The absence of correlation between markers of neoangiogenesis and (18)FDG uptake suggests a temporal separation between the process of neoangiogenesis and inflammatory activity.
Collapse
Affiliation(s)
- Sune Folke Pedersen
- Cluster for Molecular Imaging, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
46
|
Fogg VC, Lanning NJ, Mackeigan JP. Mitochondria in cancer: at the crossroads of life and death. CHINESE JOURNAL OF CANCER 2012; 30:526-39. [PMID: 21801601 PMCID: PMC3336361 DOI: 10.5732/cjc.011.10018] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondrial processes play an important role in tumor initiation and progression. In this review, we focus on three critical processes by which mitochondrial function may contribute to cancer: through alterations in glucose metabolism, the production of reactive oxygen species (ROS) and compromise of intrinsic apoptotic function. Alterations in cancer glucose metabolism include the Warburg effect, leading to a shift in metabolism away from aerobic respiration toward glycolysis, even when sufficient oxygen is present to support respiration. Such alterations in cellular metabolism may favor tumor cell growth by increasing the availability of biosynthetic intermediates needed for cellular growth and proliferation. Mutations in specific metabolic enzymes, namely succinate dehydrogenase, fumarate hydratase and the isocitrate dehydrogenases, have been linked to human cancer. Mitochondrial ROS may contribute to cancer via DNA damage and the activation of aberrant signaling pathways. ROS-dependent stabilization of the transcription factor hypoxia-inducible factor (HIF) may be a particularly important event for tumorigenesis. Compromised function of intrinsic apoptosis removes an important cellular safeguard against cancer and has been implicated in tumorigenesis, tumor metastasis, and chemoresistance. Each of the major mitochondrial processes is linked. In this review, we outline the connections between them and address ways these mitochondrial pathways may be targeted for cancer therapy.
Collapse
Affiliation(s)
- Vanessa C Fogg
- Laboratory of Systems Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | | |
Collapse
|
47
|
Rau TF, Lu Q, Sharma S, Sun X, Leary G, Beckman ML, Hou Y, Wainwright MS, Kavanaugh M, Poulsen DJ, Black SM. Oxygen glucose deprivation in rat hippocampal slice cultures results in alterations in carnitine homeostasis and mitochondrial dysfunction. PLoS One 2012; 7:e40881. [PMID: 22984394 PMCID: PMC3439445 DOI: 10.1371/journal.pone.0040881] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 06/18/2012] [Indexed: 12/02/2022] Open
Abstract
Mitochondrial dysfunction characterized by depolarization of mitochondrial membranes and the initiation of mitochondrial-mediated apoptosis are pathological responses to hypoxia-ischemia (HI) in the neonatal brain. Carnitine metabolism directly supports mitochondrial metabolism by shuttling long chain fatty acids across the inner mitochondrial membrane for beta-oxidation. Our previous studies have shown that HI disrupts carnitine homeostasis in neonatal rats and that L-carnitine can be neuroprotective. Thus, this study was undertaken to elucidate the molecular mechanisms by which HI alters carnitine metabolism and to begin to elucidate the mechanism underlying the neuroprotective effect of L-carnitine (LCAR) supplementation. Utilizing neonatal rat hippocampal slice cultures we found that oxygen glucose deprivation (OGD) decreased the levels of free carnitines (FC) and increased the acylcarnitine (AC): FC ratio. These changes in carnitine homeostasis correlated with decreases in the protein levels of carnitine palmitoyl transferase (CPT) 1 and 2. LCAR supplementation prevented the decrease in CPT1 and CPT2, enhanced both FC and the AC∶FC ratio and increased slice culture metabolic viability, the mitochondrial membrane potential prior to OGD and prevented the subsequent loss of neurons during later stages of reperfusion through a reduction in apoptotic cell death. Finally, we found that LCAR supplementation preserved the structural integrity and synaptic transmission within the hippocampus after OGD. Thus, we conclude that LCAR supplementation preserves the key enzymes responsible for maintaining carnitine homeostasis and preserves both cell viability and synaptic transmission after OGD.
Collapse
Affiliation(s)
- Thomas F. Rau
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana, United States of America
| | - Qing Lu
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Shruti Sharma
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Xutong Sun
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Gregory Leary
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana, United States of America
| | - Matthew L. Beckman
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana, United States of America
| | - Yali Hou
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Mark S. Wainwright
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Children's Memorial Hospital, 2300 Children's Plaza, Chicago, Illinois, United States of America
| | - Michael Kavanaugh
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana, United States of America
| | - David J. Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, Montana, United States of America
- * E-mail: (SMB); (DJP)
| | - Stephen M. Black
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, United States of America
- * E-mail: (SMB); (DJP)
| |
Collapse
|
48
|
Emerging glycolysis targeting and drug discovery from chinese medicine in cancer therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:873175. [PMID: 22844340 PMCID: PMC3403522 DOI: 10.1155/2012/873175] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 05/28/2012] [Accepted: 06/12/2012] [Indexed: 02/04/2023]
Abstract
Molecular-targeted therapy has been developed for cancer chemoprevention and treatment. Cancer cells have different metabolic properties from normal cells. Normal cells mostly rely upon the process of mitochondrial oxidative phosphorylation to produce energy whereas cancer cells have developed an altered metabolism that allows them to sustain higher proliferation rates. Cancer cells could predominantly produce energy by glycolysis even in the presence of oxygen. This alternative metabolic characteristic is known as the “Warburg Effect.” Although the exact mechanisms underlying the Warburg effect are unclear, recent progress indicates that glycolytic pathway of cancer cells could be a critical target for drug discovery. With a long history in cancer treatment, traditional Chinese medicine (TCM) is recognized as a valuable source for seeking bioactive anticancer compounds. A great progress has been made to identify active compounds from herbal medicine targeting on glycolysis for cancer treatment. Herein, we provide an overall picture of the current understanding of the molecular targets in the cancer glycolytic pathway and reviewed active compounds from Chinese herbal medicine with the potentials to inhibit the metabolic targets for cancer treatment. Combination of TCM with conventional therapies will provide an attractive strategy for improving clinical outcome in cancer treatment.
Collapse
|
49
|
McIntyre DJO, Madhu B, Lee SH, Griffiths JR. Magnetic resonance spectroscopy of cancer metabolism and response to therapy. Radiat Res 2012; 177:398-435. [PMID: 22401303 DOI: 10.1667/rr2903.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Magnetic resonance spectroscopy allows noninvasive in vivo measurements of biochemical information from living systems, ranging from cultured cells through experimental animals to humans. Studies of biopsies or extracts offer deeper insights by detecting more metabolites and resolving metabolites that cannot be distinguished in vivo. The pharmacokinetics of certain drugs, especially fluorinated drugs, can be directly measured in vivo. This review briefly describes these methods and their applications to cancer metabolism, including glycolysis, hypoxia, bioenergetics, tumor pH, and tumor responses to radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Dominick J O McIntyre
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| | | | | | | |
Collapse
|
50
|
Kon M, Kiffin R, Koga H, Chapochnick J, Macian F, Varticovski L, Cuervo AM. Chaperone-mediated autophagy is required for tumor growth. Sci Transl Med 2012; 3:109ra117. [PMID: 22089453 DOI: 10.1126/scitranslmed.3003182] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cellular process of autophagy (literally "self-eating") is important for maintaining the homeostasis and bioenergetics of mammalian cells. Two of the best-studied mechanisms of autophagy are macroautophagy and chaperone-mediated autophagy (CMA). Changes in macroautophagy activity have been described in cancer cells and in solid tumors, and inhibition of macroautophagy promotes tumorigenesis. Because normal cells respond to inhibition of macroautophagy by up-regulation of the CMA pathway, we aimed to characterize the CMA status in different cancer cells and to determine the contribution of changes in CMA to tumorigenesis. Here, we show consistent up-regulation of CMA in different types of cancer cells regardless of the status of macroautophagy. We also demonstrate an increase in CMA components in human cancers of different types and origins. CMA is required for cancer cell proliferation in vitro because it contributes to the maintenance of the metabolic alterations characteristic of malignant cells. Using human lung cancer xenografts in mice, we confirmed the CMA dependence of cancer cells in vivo. Inhibition of CMA delays xenograft tumor growth, reduces the number of cancer metastases, and induces regression of existing human lung cancer xenografts in mice. The fact that similar manipulations of CMA also reduce tumor growth of two different melanoma cell lines suggests that targeting this autophagic pathway may have broad antitumorigenic potential.
Collapse
Affiliation(s)
- Maria Kon
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|