601
|
Somasundaram R, Herlyn M, Wagner SN. The role of tumor microenvironment in melanoma therapy resistance. Melanoma Manag 2016; 3:23-32. [PMID: 30190870 PMCID: PMC6094607 DOI: 10.2217/mmt.15.37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Melanoma patients develop resistance to both chemotherapy and targeted-therapy drugs. Promising preclinical and clinical results with immune checkpoint inhibitors using antibodies directed against cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 have re-energized the field of immune-based therapies in melanoma. However, similar to chemotherapy or targeted therapies, immune checkpoint blockade responds in only subsets of melanoma patients. A number of factors, including gene mutations, altered cell-signaling pathways and tumor heterogeneity can contribute to therapy resistance. Recent studies have highlighted the role of inflammatory tumor microenvironment on therapy resistance of cancer cells. Cancer cells either alone or in conjunction with the tumor stroma can contribute to an inflammatory microenvironment. Multimodal approaches of targeting the tumor microenvironment, in addition to malignant cells, may be necessary for better therapy responses.
Collapse
Affiliation(s)
| | - Meenhard Herlyn
- The Wistar Institute, 3601 Spruce St, Philadelphia, PA 19104, USA
| | - Stephan N Wagner
- Division of Immunology, Allergy & Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Wien, Austria
| |
Collapse
|
602
|
Gu Q, He Y, Ji J, Yao Y, Shen W, Luo J, Zhu W, Cao H, Geng Y, Xu J, Zhang S, Cao J, Ding WQ. Hypoxia-inducible factor 1α (HIF-1α) and reactive oxygen species (ROS) mediates radiation-induced invasiveness through the SDF-1α/CXCR4 pathway in non-small cell lung carcinoma cells. Oncotarget 2016; 6:10893-907. [PMID: 25843954 PMCID: PMC4484427 DOI: 10.18632/oncotarget.3535] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/18/2015] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is an important procedure for the treatment of inoperable non-small cell lung cancer (NSCLC). However, recent evidence has shown that irradiation can promote the invasion and metastasis of several types of cancer, and the underlying mechanisms are not fully understood. This study aimed to investigate the molecular mechanism by which radiation enhances the invasiveness of NSCLC cells. We found that after irradiation, hypoxia-inducible factor 1α (HIF-1α) was increased and translocated into the nucleus, where it bound to the hypoxia response element (HRE) in the CXCR4 promoter and promoted the transcription of CXCR4. Furthermore, reactive oxygen species (ROS) also plays a role in the radiation-induced expression of CXCR4. Our results revealed that 2 Gy X-ray irradiation promoted the metastasis and invasiveness of H1299, A549 and H460 cells, which were significantly enhanced by SDF-1α treatment. Blocking the SDF-1α/CXCR4 interaction could suppress the radiation-induced invasiveness of NSCLC cells. The PI3K/pAkt and MAPK/pERK1/2 pathways were found to be involved in radiation-induced matrix metalloproteinase (MMP) expression. In vivo, irradiation promoted the colonization of H1299 cells in the liver and lung, which was mediated by CXCR4. Altogether, our findings have elucidated the underlying mechanisms of the irradiation-enhanced invasiveness of NSCLC cells.
Collapse
Affiliation(s)
- Qing Gu
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China.,Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yan He
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Jianfeng Ji
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, China
| | - Yifan Yao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Wenhao Shen
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Jialin Luo
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Zhu
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Han Cao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Yangyang Geng
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Jing Xu
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Shuyu Zhang
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Jianping Cao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China.,Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, China
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma, United States
| |
Collapse
|
603
|
Zhao Y, Wang X, Li L, Li C. Doxycycline inhibits proliferation and induces apoptosis of both human papillomavirus positive and negative cervical cancer cell lines. Can J Physiol Pharmacol 2016; 94:526-33. [PMID: 26913972 DOI: 10.1139/cjpp-2015-0481] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The clinical management of cervical cancer remains a challenge and the development of new treatment strategies merits attention. However, the discovery and development of novel compounds can be a long and labourious process. Drug repositioning may circumvent this process and facilitate the rapid translation of hypothesis-driven science into the clinics. In this work, we show that a FDA-approved antibiotic, doxycycline, effectively targets human papillomavirus (HPV) positive and negative cervical cancer cells in vitro and in vivo. Doxycycline significantly inhibits proliferation of a panel of cervical cancer cell lines. It also induces apoptosis of cervical cancer cells in a time- and dose-dependent manner. In addition, the apoptosis induced by doxycycline is through caspase-dependent pathway. Mechanism studies demonstrate that doxycycline affects oxygen consumption rate, glycolysis, and reduces ATP levels in cervical cancer cells. In HeLa xenograft mouse model, doxycycline significantly inhibits growth of tumour. Our in vitro and in vivo data clearly demonstrate the inhibitory effects of doxycycline on the growth and survival of cervical cancer cells. Our work provides the evidence that doxycycline can be repurposed for the treatment of cervical cancer and targeting energy metabolism may represent a potential therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Yan Zhao
- Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China
| | - Xinyu Wang
- Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China
| | - Lei Li
- Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China.,Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China
| | - Changzhong Li
- Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China.,Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwu Rd., Huaiyin Dist., Jinan 250021, Shandong Province, China
| |
Collapse
|
604
|
Tang XY, Zheng W, Ding M, Guo KJ, Yuan F, Feng H, Deng B, Sun W, Hou Y, Gao L. miR-125b acts as a tumor suppressor in chondrosarcoma cells by the sensitization to doxorubicin through direct targeting the ErbB2-regulated glucose metabolism. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:571-83. [PMID: 26966351 PMCID: PMC4771406 DOI: 10.2147/dddt.s90530] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chondrosarcoma is the second most common type of primary bone malignancy in the United States after osteosarcoma. Surgical resections of these tumors are the only effective treatment to chondrosarcoma patients due to their resistance to conventional chemo- and radiotherapy. In this study, miR-125b was found to perform its tumor-suppressor function to inhibit glucose metabolism via the direct targeting of oncogene, ErbB2. We report miR-125b was downregulated in both chondrosarcoma patient samples and cell lines. The total 20 Asian chondrosarcoma patients showed significantly downregulated miR-125b expression compared with normal tissues. Meanwhile, miR-125 was downregulated in chondrosarcoma cells and doxorubicin resistant cells. Overexpression of miR-125 enhanced the sensitivity of both parental and doxorubicin resistant cells to doxorubicin through direct targeting on the ErbB2-mediated upregulation of glycolysis in chondrosarcoma cells. Moreover, restoration of the expression of ErbB2 and glucose metabolic enzymes in miR-125 pretransfected cells recovered the susceptibility to doxorubicin. Our study will provide a novel aspect on the overcoming chemoresistance in human chondrosarcoma cells and may help in the development of therapeutic strategies for the treatments of patients.
Collapse
Affiliation(s)
- Xian-ye Tang
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China; Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Wei Zheng
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China; Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Min Ding
- Department of Emergency Trauma Surgery, East Hospital Affiliated to Tongji University
| | - Kai-jin Guo
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China; Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Feng Yuan
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hu Feng
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China
| | - Bin Deng
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Sun
- Department of Orthopedic, The First Clinical Medical College, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yang Hou
- Department of Orthopedic, Changzheng Hospital, Shanghai, People's Republic of China
| | - Lu Gao
- College of Life Sciences, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China; Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
605
|
Gamberi T, Magherini F, Fiaschi T, Landini I, Massai L, Valocchia E, Bianchi L, Bini L, Gabbiani C, Nobili S, Mini E, Messori L, Modesti A. Proteomic analysis of the cytotoxic effects induced by the organogold(III) complex Aubipyc in cisplatin-resistant A2780 ovarian cancer cells: further evidence for the glycolytic pathway implication. MOLECULAR BIOSYSTEMS 2016; 11:1653-67. [PMID: 25906354 DOI: 10.1039/c5mb00008d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The cellular alterations produced in cisplatin-resistant A2780 ovarian cancer cells (A2780/R) upon treatment with the cytotoxic organogold(III) complex Aubipyc were investigated in depth through a classical proteomic approach. We observed that A2780/R cell exposure to a cytotoxic concentration of Aubipyc for 24 hours results in a conspicuous number of alterations at the protein level that were carefully examined. Notably, we observed that several affected proteins belong to the glucose metabolism system further supporting the idea that the cytotoxic effects of Aubipyc in A2780/R cells are mostly mediated by an impairment of glucose metabolism in excellent agreement with previous observations on the parent cisplatin-sensitive cell line.
Collapse
Affiliation(s)
- Tania Gamberi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
606
|
Blockage of glutaminolysis enhances the sensitivity of ovarian cancer cells to PI3K/mTOR inhibition involvement of STAT3 signaling. Tumour Biol 2016; 37:11007-15. [PMID: 26894601 DOI: 10.1007/s13277-016-4984-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/07/2016] [Indexed: 12/30/2022] Open
Abstract
The PI3K/Akt/mTOR axis in ovarian cancer is frequently activated and implicated in tumorigenesis. Specific targeting of this pathway is therefore an attractive therapeutic approach for ovarian cancer. However, ovarian cancer cells are resistant to PP242, a dual inhibitor of mTORC1 and mTORC2. Interestingly, blockage of GLS1 with a selective inhibitor, CB839, or siRNA dramatically sensitized the PP242-induced cell death, as evident from increased PARP cleavage. The anti-cancer activity of CB-839 and PP242 was abrogated by the addition of the TCA cycle product α-ketoglutarate, indicating the critical function of GLS1 in ovarian cancer cell survival. Finally, glutaminolysis inhibition activated apoptosis and synergistically sensitized ovarian cancer cells to priming with the mTOR inhibitor PP242. GLS1 inhibition significantly reduced phosphorylated STAT3 expression in ovarian cancer cells. These findings show that targeting glutamine addiction via GLS1 inhibition offers a potential novel therapeutic strategy to overcome resistance to PI3K/Akt/mTOR inhibition.
Collapse
|
607
|
Glycolytic metabolism influences global chromatin structure. Oncotarget 2016; 6:4214-25. [PMID: 25784656 PMCID: PMC4414184 DOI: 10.18632/oncotarget.2929] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/15/2014] [Indexed: 12/20/2022] Open
Abstract
Metabolic rewiring, specifically elevated glycolytic metabolism is a hallmark of cancer. Global chromatin structure regulates gene expression, DNA repair, and also affects cancer progression. But the interrelationship between tumor metabolism and chromatin architecture remain unclear. Here we show that increased glycolysis in cancer cells promotes an open chromatin configuration. Using complementary methods including Micrococcal nuclease (MNase) digestion assay, electron microscope and immunofluorescence staining, we demonstrate that glycolysis inhibition by pharmacological and genetic approaches was associated with induction of compacted chromatin structure. This condensed chromatin status appeared to result chiefly from histone hypoacetylation as restoration of histone acetylation with an HDAC inhibitor reversed the compacted chromatin state. Interestingly, glycolysis inhibition-induced chromatin condensation impeded DNA repair efficiency leading to increased sensitivity of cancer cells to DNA damage drugs, which may represent a novel molecular mechanism that can be exploited for cancer therapy.
Collapse
|
608
|
Li Volsi A, Jimenez de Aberasturi D, Henriksen-Lacey M, Giammona G, Licciardi M, Liz-Marzán LM. Inulin coated plasmonic gold nanoparticles as a tumor-selective tool for cancer therapy. J Mater Chem B 2016; 4:1150-1155. [PMID: 28261481 PMCID: PMC5317210 DOI: 10.1039/c5tb01810b] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/02/2016] [Indexed: 11/28/2022]
Abstract
Polymer coated gold nanospheres are proposed as a tumor selective carrier for the anticancer drug doxorubicin. Thiolated polyethyleneglycol (PEG-SH) and an inulin-amino derivative based copolymer (INU-EDA) were used as stabilizing and coating materials for 40 nm gold nanospheres. The resulting polymer coated gold nanospheres (Au@PEG-INU) showed excellent physicochemical stability and potential stealth like behavior. The system was loaded with doxorubicin (Au@PEG-INU/Doxo) and its cytotoxicity profile was evaluated on human cervical cancer cells (HeLa) and lung cancer cells (A549), as compared to Au@PEG-INU and doxorubicin alone. Cytotoxicity assays showed that the system is able to drastically reduce cell viability upon incubation for 3 days. This result was supported by the ability of Au@PEG-INU/Doxo to be internalized by cancer cells and to release doxorubicin, as assessed by fluorescence microscopy. Finally, a cancer/non cancer cell co-culture model was used to display the advantageous therapeutic effects of the proposed system with respect to doxorubicin alone, thereby demonstrating the ability of Au@PEG-INU/Doxo to preferentially accumulate in tumor cells due to their enhanced metabolism, and to selectively kill target cells.
Collapse
Affiliation(s)
- Anna Li Volsi
- Laboratory of Biocompatible Polymers , Department of Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF) , University of Palermo , Via Archirafi, 32 , 90123 Palermo , Italy . ; ; Tel: +39 091 23891927
| | | | - Malou Henriksen-Lacey
- Bionanoplasmonics Laboratory , CIC biomaGUNE , Paseo de Miramón 182 , 20009 Donostia San-Sebastian , Spain .
| | - Gaetano Giammona
- Laboratory of Biocompatible Polymers , Department of Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF) , University of Palermo , Via Archirafi, 32 , 90123 Palermo , Italy . ; ; Tel: +39 091 23891927 ; Mediterranean Center for Human Health Advanced Biotechnologies (Med-CHAB) , Palermo , Italy
| | - Mariano Licciardi
- Laboratory of Biocompatible Polymers , Department of Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF) , University of Palermo , Via Archirafi, 32 , 90123 Palermo , Italy . ; ; Tel: +39 091 23891927 ; Mediterranean Center for Human Health Advanced Biotechnologies (Med-CHAB) , Palermo , Italy
| | - Luis M Liz-Marzán
- Bionanoplasmonics Laboratory , CIC biomaGUNE , Paseo de Miramón 182 , 20009 Donostia San-Sebastian , Spain . ; Ikerbasque , Basque Foundation for Science , 48013 Bilbao , Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN) , Paseo de Miramón 182 , 20009 Donostia-San Sebastian , Spain
| |
Collapse
|
609
|
Shinohara H, Kumazaki M, Minami Y, Ito Y, Sugito N, Kuranaga Y, Taniguchi K, Yamada N, Otsuki Y, Naoe T, Akao Y. Perturbation of energy metabolism by fatty-acid derivative AIC-47 and imatinib in BCR-ABL-harboring leukemic cells. Cancer Lett 2016; 371:1-11. [DOI: 10.1016/j.canlet.2015.11.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 12/18/2022]
|
610
|
Glutamine drives glutathione synthesis and contributes to radiation sensitivity of A549 and H460 lung cancer cell lines. Biochim Biophys Acta Gen Subj 2016; 1860:836-43. [PMID: 26825773 DOI: 10.1016/j.bbagen.2016.01.021] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/09/2015] [Accepted: 01/24/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increased glutamine uptake is known to drive cancer cell proliferation, making tumor cells glutamine-dependent. Glutamine provides additional carbon and nitrogen sources for cell growth. The first step in glutamine utilization is its conversion to glutamate by glutaminase (GLS). Glutamate is a precursor for glutathione synthesis, and we investigated the hypothesis that glutamine drives glutathione synthesis and thereby contributes to cellular defense systems. METHODS The importance of glutamine for glutathione synthesis was studied in H460 and A549 lung cancer cell lines using glutamine-free medium and bis-2-(5-phenyl-acetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) a GLS inhibitor. Metabolic activities were determined by targeted mass spectrometry. RESULTS A significant correlation between glutamine consumption and glutathione excretion was demonstrated in H460 and A549 tumor cells. Culturing in the presence of [(13)C5]glutamine demonstrated that by 12h >50% of excreted glutathione was derived from glutamine. Culturing in glutamine-free medium or treatment with BPTES, a GLS-specific inhibitor, reduced cell proliferation and viability and abolished glutathione excretion. Treatment with glutathione-ester prevented BPTES-induced cytotoxicity. Inhibition of GLS markedly radiosensitized the lung tumor cell lines, suggesting an important role of glutamine-derived glutathione in determining radiation sensitivity. CONCLUSIONS We demonstrate here for the first time that a significant amount of extracellular glutathione is directly derived from glutamine. This finding adds yet another important function to the already known glutamine dependence of tumor cells and probably tumors as well. GENERAL SIGNIFICANCE Glutamine is essential for synthesis and excretion of glutathione to promote cell growth and viability.
Collapse
|
611
|
Bacci M, Giannoni E, Fearns A, Ribas R, Gao Q, Taddei ML, Pintus G, Dowsett M, Isacke CM, Martin LA, Chiarugi P, Morandi A. miR-155 Drives Metabolic Reprogramming of ER+ Breast Cancer Cells Following Long-Term Estrogen Deprivation and Predicts Clinical Response to Aromatase Inhibitors. Cancer Res 2016; 76:1615-26. [DOI: 10.1158/0008-5472.can-15-2038] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/28/2015] [Indexed: 11/16/2022]
|
612
|
Pópulo H, Caldas R, Lopes JM, Pardal J, Máximo V, Soares P. Overexpression of pyruvate dehydrogenase kinase supports dichloroacetate as a candidate for cutaneous melanoma therapy. Expert Opin Ther Targets 2016; 19:733-45. [PMID: 25976231 DOI: 10.1517/14728222.2015.1045416] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE We aimed to verify if there is evidence to consider dichloroacetate (DCA), which inhibits the pyruvate dehydrogenase kinase (PDK) and reverts the metabolic shift of cancer cells from glycolysis to oxidative phosphorylation, as a promising drug for therapy of cutaneous melanoma (CM) patients. RESEARCH DESIGN AND METHODS We assessed the expression profile of PDK 1, 2 and 3 in a series of melanoma samples, to verify if melanoma tumors express the DCA targets, if this expression correlates with the activation of important signaling cascades for melanomagenesis and also with the prognosis of melanoma patients. We also established the sensitivity of melanoma cell lines to DCA treatment, by assessing their metabolic alterations, proliferation and survival. RESULTS We observed that both PDK 1 and 2 isoforms are overexpressed in CM compared to nevi, this expression being associated with the expression of the mTOR pathway effectors and independent of the BRAF mutational status. Melanoma cell lines treated with DCA showed a shift in metabolism, that is, a decrease in glucose consumption and lactate production, downregulation of proliferation, an increase of apoptosis and a decrease in mTOR pathway activation. CONCLUSION Our results suggest that PDK expression may play a role in melanoma development and that DCA can be useful for CM therapy, alone or in combination with mTOR inhibitors.
Collapse
Affiliation(s)
- Helena Pópulo
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), University of Porto , Porto , Portugal +22 557 0700 ; +22 557 0799 ;
| | | | | | | | | | | |
Collapse
|
613
|
Genetic alterations in fatty acid transport and metabolism genes are associated with metastatic progression and poor prognosis of human cancers. Sci Rep 2016; 6:18669. [PMID: 26725848 PMCID: PMC4698658 DOI: 10.1038/srep18669] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/17/2015] [Indexed: 01/18/2023] Open
Abstract
Reprogramming of cellular metabolism is a hallmark feature of cancer cells. While a distinct set of processes drive metastasis when compared to tumorigenesis, it is yet unclear if genetic alterations in metabolic pathways are associated with metastatic progression of human cancers. Here, we analyzed the mutation, copy number variation and gene expression patterns of a literature-derived model of metabolic genes associated with glycolysis (Warburg effect), fatty acid metabolism (lipogenesis, oxidation, lipolysis, esterification) and fatty acid uptake in >9000 primary or metastatic tumor samples from the multi-cancer TCGA datasets. Our association analysis revealed a uniform pattern of Warburg effect mutations influencing prognosis across all tumor types, while copy number alterations in the electron transport chain gene SCO2, fatty acid uptake (CAV1, CD36) and lipogenesis (PPARA, PPARD, MLXIPL) genes were enriched in metastatic tumors. Using gene expression profiles, we established a gene-signature (CAV1, CD36, MLXIPL, CPT1C, CYP2E1) that strongly associated with epithelial-mesenchymal program across multiple cancers. Moreover, stratification of samples based on the copy number or expression profiles of the genes identified in our analysis revealed a significant effect on patient survival rates, thus confirming prominent roles of fatty acid uptake and metabolism in metastatic progression and poor prognosis of human cancers.
Collapse
|
614
|
Ghosh M, Saha S, Dutta SK. Synergism of metabolic modulators Bet-CA and LDCA: a rational combinatorial approach to selectively combat cancer associated hallmark traits. RSC Adv 2016. [DOI: 10.1039/c6ra14366k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The strategic combination of two metabolic modulators, Bet-CA and LDCA in a calculated ratio selectively and synergistically inhibits the hallmark traits of cancer by altering the highly resilient mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Monisankar Ghosh
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Suchandrima Saha
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Samir Kumar Dutta
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| |
Collapse
|
615
|
Zhang J, Park D, Shin DM, Deng X. Targeting KRAS-mutant non-small cell lung cancer: challenges and opportunities. Acta Biochim Biophys Sin (Shanghai) 2016; 48:11-6. [PMID: 26578706 DOI: 10.1093/abbs/gmv118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022] Open
Abstract
Oncogenic mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) occur in 15%-30% of non-small cell lung cancer (NSCLC). However, despite decades of intensive research, there is still no direct KRAS inhibitor with clinically proven efficacy. Considering its association with poor treatment response and prognosis of lung cancer, developing an effective inhibitory approach is urgently needed. Here, we review different strategies currently being explored to target KRAS-mutant NSCLC, discuss opportunities and challenges, and also propose some novel methods and concepts with the promise of clinical application.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Dongkyoo Park
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Dong M Shin
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
616
|
Qiu P, Man S, Yang H, Liu Y, Liu Z, Ma L, Yu P, Gao W. Metabolic regulatory network alterations reveal different therapeutic effects of cisplatin and Rhizoma paridis saponins in Lewis pulmonary adenoma mice. RSC Adv 2016. [DOI: 10.1039/c6ra23382a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Metabonomics is used to compare the metabolic profiling of RPS and DDP in Lewis pulmonary adenoma mice; RPS is found to be a potent anticancer agent through inhibiting cancer cellular metabolism to suppress metastases in murine lung adenocarcinoma.
Collapse
Affiliation(s)
- Peiyu Qiu
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - Shuli Man
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - He Yang
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - Yuanxue Liu
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency
- School of Pharmaceutical Science and Technology
- Tianjin University
- Tianjin
- China
| | - Zhen Liu
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - Long Ma
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - Peng Yu
- Key Laboratory of Industrial Microbiology
- Ministry of Education
- Tianjin Key Laboratory of Industry Microbiology
- National and Local United Engineering Lab of Metabolic Control Fermentation Technology
- College of Biotechnology
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency
- School of Pharmaceutical Science and Technology
- Tianjin University
- Tianjin
- China
| |
Collapse
|
617
|
Gao S, Chen X, Jin H, Ren S, Liu Z, Fang X, Zhang G. Overexpression of ErbB2 renders breast cancer cells susceptible to 3-BrPA through the increased dissociation of hexokinase II from mitochondrial outer membrane. Oncol Lett 2015; 11:1567-1573. [PMID: 26893781 DOI: 10.3892/ol.2015.4043] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/16/2015] [Indexed: 01/05/2023] Open
Abstract
ErbB2 is known to upregulate glycolysis in breast cancer, however, the precise mechanisms remain unclear. In the present study, ErbB2 upregulated Hexokinase II (HK II) activity by increasing the binding of HK II to the mitochondrial outer membrane. Dysregulated glucose metabolism in high ErbB2-expressing breast cancer cells induces susceptibility to glucose starvation and glycolysis inhibition. Additionally, HK II has a tendency to dissociate from the mitochondria outer membrane in ErbB2-overexpressing cells following treatment with the HK II inhibitor, 3-BrPA. Furthermore, 3-BrPA treatment results in decreased mitochondria membrane potential and release of cytochrome c into cytoplasm in ErbB2-overexpressing cells, leading to activation of the mitochondrial apoptotic signaling pathway. In summary, the results demonstrate a novel mechanism for ErbB2-activated glycolysis and reveal that 3-BrPA is effective in reducing ErbB2-positive breast cancer cell viability by targeting HK II in vitro and in vivo.
Collapse
Affiliation(s)
- Sujie Gao
- Department of Anesthesiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China; Central Research Laboratory, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xuebo Chen
- Department of General Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hongyong Jin
- Department of General Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Shengnan Ren
- Department of General Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhuo Liu
- Department of General Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xuedong Fang
- Department of General Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guizhen Zhang
- Central Research Laboratory, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
618
|
Mocanu MM, Nagy P, Szöllősi J. Chemoprevention of Breast Cancer by Dietary Polyphenols. Molecules 2015; 20:22578-620. [PMID: 26694341 PMCID: PMC6332464 DOI: 10.3390/molecules201219864] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
The review will discuss in detail the effects of polyphenols on breast cancer, including both the advantages and disadvantages of the applications of these natural compounds. First, we focus on the characterization of the main classes of polyphenols and then on in vitro and in vivo experiments carried out in breast cancer models. Since the therapeutic effects of the administration of a single type of polyphenol might be limited because of the reduced bioavailability of these drugs, investigations on combination of several polyphenols or polyphenols with conventional therapy will also be discussed. In addition, we present recent data focusing on clinical trials with polyphenols and new approaches with nanoparticles in breast cancer. Besides the clinical and translational findings this review systematically summarizes our current knowledge about the molecular mechanisms of anti-cancer effects of polyphenols, which are related to apoptosis, cell cycle regulation, plasma membrane receptors, signaling pathways and epigenetic mechanisms. At the same time the effects of polyphenols on primary tumor, metastasis and angiogenesis in breast cancer are discussed. The increasing enthusiasm regarding the combination of polyphenols and conventional therapy in breast cancer might lead to additional efforts to motivate further research in this field.
Collapse
Affiliation(s)
- Maria-Magdalena Mocanu
- Department of Biophysics, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
619
|
Nevedomskaya E, Perryman R, Solanki S, Syed N, Mayboroda OA, Keun HC. A Systems Oncology Approach Identifies NT5E as a Key Metabolic Regulator in Tumor Cells and Modulator of Platinum Sensitivity. J Proteome Res 2015; 15:280-90. [PMID: 26629888 DOI: 10.1021/acs.jproteome.5b00793] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Altered metabolism in tumor cells is required for rapid proliferation but also can influence other phenotypes that affect clinical outcomes such as metastasis and sensitivity to chemotherapy. Here, a genome-wide association study (GWAS)-guided integration of NCI-60 transcriptome and metabolome data identified ecto-5'-nucleotidase (NT5E or CD73) as a major determinant of metabolic phenotypes in cancer cells. NT5E expression and associated metabolome variations were also correlated with sensitivity to several chemotherapeutics including platinum-based treatment. NT5E mRNA levels were observed to be elevated in cells upon in vitro and in vivo acquisition of platinum resistance in ovarian cancer cells, and specific targeting of NT5E increased tumor cell sensitivity to platinum. We observed that tumor NT5E levels were prognostic for outcomes in ovarian cancer and were elevated after treatment with platinum, supporting the translational relevance of our findings. In this work, we integrated and analyzed a plethora of public data, demonstating the merit of such a systems oncology approach for the discovery of novel players in cancer biology and therapy. We experimentally validated the main findings of the NT5E gene being involved in both intrinsic and acquired resistance to platinum-based drugs. We propose that the efficacy of conventional chemotherapy could be improved by NT5E inhibition and that NT5E expression may be a useful prognostic and predictive clinical biomarker.
Collapse
Affiliation(s)
- Ekaterina Nevedomskaya
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC) , L4-Q, PO Box 9600, 2300RC Leiden, The Netherlands
| | - Richard Perryman
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital , London W12 0NN, United Kingdom.,Division of Brain Sciences, Department of Medicine, Imperial College London , London W12 0NN, United Kingdom
| | - Shyam Solanki
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital , London W12 0NN, United Kingdom
| | - Nelofer Syed
- Division of Brain Sciences, Department of Medicine, Imperial College London , London W12 0NN, United Kingdom
| | - Oleg A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC) , L4-Q, PO Box 9600, 2300RC Leiden, The Netherlands
| | - Hector C Keun
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital , London W12 0NN, United Kingdom
| |
Collapse
|
620
|
Cort A, Ozben T, Melchiorre M, Chatgilialoglu C, Ferreri C, Sansone A. Effects of bleomycin and antioxidants on the fatty acid profile of testicular cancer cell membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:434-41. [PMID: 26656160 DOI: 10.1016/j.bbamem.2015.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/01/2015] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
Abstract
Bleomycin is used in chemotherapy regimens for the treatment of patients having testicular germ-cell tumor (TGCT). There is no study in the literature investigating the effects of bleomycin on membrane lipid profile in testicular cancer cells. We investigated membrane fatty acid (FA) profiles isolated, derivatized and analyzed by gas chromatography of NTera-2 testicular cancer cells incubated with bleomycin (Bleo) for 24 h in the absence and presence of N-Acetyl-L-Cysteine (NAC) and curcumin (Cur) as commonly used antioxidant adjuvants. At the same time the MAPK pathway and EGFR levels were followed up. Bleomycin treatment increased significantly saturated fatty acids (SFA) of phospholipids at the expense of monounsaturated (MUFA) and polyunsaturated fatty acids (PUFA). Bleomycin also led to a significant increase in the trans lipid isomers of oleic and arachidonic acids due to its free radical producing effect. Incubation with bleomycin increased the p38 MAPK and JNK levels and downregulated EGFR pathway. Coincubation of bleomycin with NAC reversed effects caused by bleomycin. Our results highlight the important role of membrane fatty acid remodeling occurring during the use of bleomycin and its concurrent use with antioxidants which can adjuvate the cytotoxic effects of the chemotherapeutic agents.
Collapse
Affiliation(s)
- A Cort
- Department of Medical Biochemistry, School of Medicine, Faculty of Health Sciences SANKO University, Gaziantep, Turkey; Department of Nutrition and Dietetics, Faculty of Health Sciences SANKO University, Gaziantep, Turkey; Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy; Akdeniz University, Medical Faculty, Department of Biochemistry, Antalya, Turkey
| | - T Ozben
- Akdeniz University, Medical Faculty, Department of Biochemistry, Antalya, Turkey.
| | - M Melchiorre
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - C Chatgilialoglu
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", Aghia Paraskevi, Athens, Greece
| | - C Ferreri
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - A Sansone
- Institute for the Organic Synthesis and Photoreactivity, Consiglio Nazionale delle Ricerche, Bologna, Italy
| |
Collapse
|
621
|
Bhute VJ, Palecek SP. Metabolic responses induced by DNA damage and poly (ADP-ribose) polymerase (PARP) inhibition in MCF-7 cells. Metabolomics 2015; 11:1779-1791. [PMID: 26478723 PMCID: PMC4606886 DOI: 10.1007/s11306-015-0831-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 07/13/2015] [Indexed: 11/30/2022]
Abstract
Genomic instability is one of the hallmarks of cancer. Several chemotherapeutic drugs and radiotherapy induce DNA damage to prevent cancer cell replication. Cells in turn activate different DNA damage response (DDR) pathways to either repair the damage or induce cell death. These DDR pathways also elicit metabolic alterations which can play a significant role in the proper functioning of the cells. The understanding of these metabolic effects resulting from different types of DNA damage and repair mechanisms is currently lacking. In this study, we used NMR metabolomics to identify metabolic pathways which are altered in response to different DNA damaging agents. By comparing the metabolic responses in MCF-7 cells, we identified the activation of poly (ADP-ribose) polymerase (PARP) in methyl methanesulfonate (MMS)-induced DNA damage. PARP activation led to a significant depletion of NAD+. PARP inhibition using veliparib (ABT-888) was able to successfully restore the NAD+ levels in MMS-treated cells. In addition, double strand break induction by MMS and veliparib exhibited similar metabolic responses as zeocin, suggesting an application of metabolomics to classify the types of DNA damage responses. This prediction was validated by studying the metabolic responses elicited by radiation. Our findings indicate that cancer cell metabolic responses depend on the type of DNA damage responses and can also be used to classify the type of DNA damage.
Collapse
Affiliation(s)
- Vijesh J. Bhute
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
622
|
Tan L, Wang W, He G, Kuick RD, Gossner G, Kueck AS, Wahl H, Opipari AW, Liu JR. Resveratrol inhibits ovarian tumor growth in an in vivo mouse model. Cancer 2015; 122:722-9. [PMID: 26619367 DOI: 10.1002/cncr.29793] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Resveratrol inhibits the growth of ovarian carcinoma cells in vitro through the inhibition of glucose metabolism and the induction of both autophagy and apoptosis. In the current study, we investigated the metabolic and therapeutic effects of resveratrol in vivo. METHODS A fluorescent xenograft mouse model of ovarian cancer was used. Mice were treated with cisplatin, resveratrol, or vehicle alone. Tumor burden was assessed using whole-body imaging. The effect of resveratrol on glucose uptake in vivo was determined using micro-positron emission tomography scanning. To determine whether resveratrol could inhibit tumor regrowth, tumor-bearing mice were treated with cisplatin followed by either daily resveratrol or vehicle. Autophagic response in resected tumors taken from mice treated with resveratrol was examined by transmission electron microscopy. Glycolysis and mitochondrial respiration in ovarian tumor cells after treatment with resveratrol was assessed. RESULTS Mice treated with resveratrol and cisplatin were found to have a significantly reduced tumor burden compared with control animals (P<.001). Resveratrol-treated mice demonstrated a marked decrease in tumor uptake of glucose compared with controls. After treatment with cisplatin, "maintenance" resveratrol resulted in the suppression of tumor regrowth compared with mice receiving vehicle alone (P<.01). Tumors resected from mice treated with resveratrol exhibited autophagosomes consistent with the induction of autophagy. Treatment with resveratrol inhibited glycolytic response in ovarian tumor cells with high baseline glycolytic rates. CONCLUSIONS Treatment with resveratrol inhibits glucose uptake and has a significant antineoplastic effect in a preclinical mouse model of ovarian cancer. Resveratrol treatment suppresses tumor regrowth after therapy with cisplatin, suggesting that this agent has the potential to prolong disease-free survival. Cancer 2016;122:722-729. © 2015 American Cancer Society.
Collapse
Affiliation(s)
- Lijun Tan
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| | - Weimin Wang
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| | - Gong He
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| | - Rork D Kuick
- Department of Biostatistics, University of Michigan Health System, Ann Arbor, Michigan
| | - Gabrielle Gossner
- Department of Obstetrics and Gynecology, St. Luke's-Roosevelt Hospital, New York, New York
| | - Angela S Kueck
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, Connecticut
| | - Heather Wahl
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| | - Anthony W Opipari
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| | - J Rebecca Liu
- Department of Obstetrics and Gynecology, University of Michigan Health System, Ann Arbor, Michigan
| |
Collapse
|
623
|
Ruggieri V, Agriesti F, Scrima R, Laurenzana I, Perrone D, Tataranni T, Mazzoccoli C, Lo Muzio L, Capitanio N, Piccoli C. Dichloroacetate, a selective mitochondria-targeting drug for oral squamous cell carcinoma: a metabolic perspective of treatment. Oncotarget 2015; 6:1217-30. [PMID: 25544754 PMCID: PMC4359228 DOI: 10.18632/oncotarget.2721] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/11/2014] [Indexed: 12/28/2022] Open
Abstract
Reprogramming of metabolism is a well-established property of cancer cells that is receiving growing attention as potential therapeutic target. Oral squamous cell carcinomas (OSCC) are aggressive and drugs-resistant human tumours displaying wide metabolic heterogeneity depending on their malignant genotype and stage of development. Dichloroacetate (DCA) is a specific inhibitor of the PDH-regulator PDK proved to foster mitochondrial oxidation of pyruvate. In this study we tested comparatively the effects of DCA on three different OSCC-derived cell lines, HSC-2, HSC-3, PE15. Characterization of the three cell lines unveiled for HSC-2 and HSC-3 a glycolysis-reliant metabolism whereas PE15 accomplished an efficient mitochondrial oxidative phosphorylation. DCA treatment of the three OSCC cell lines, at pharmacological concentrations, resulted in stimulation of the respiratory activity and caused a remarkably distinctive pro-apoptotic/cytostatic effect on HSC-2 and HSC-3. This was accompanied with a large remodeling of the mitochondrial network, never documented before, leading to organelle fragmentation and with enhanced production of reactive oxygen species. The data here presented indicate that the therapeutic efficacy of DCA may depend on the specific metabolic profile adopted by the cancer cells with those exhibiting a deficient mitochondrial oxidative phosphorylation resulting more sensitive to the drug treatment.
Collapse
Affiliation(s)
- Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy
| | - Francesca Agriesti
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy
| | - Donatella Perrone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy.Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.,Laboratory of Pre-Clinical and Translational Research, IRCCS, CROB, Rionero in Vulture, Potenza, Italy.Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
624
|
JUNG KYUNGHO, LEE JINHEE, PARK JINWON, MOON SEUNGHWAN, CHO YOUNGSEOK, CHOE YEARNSEONG, LEE KYUNGHAN. Effects of curcumin on cancer cell mitochondrial function and potential monitoring with 18F-FDG uptake. Oncol Rep 2015; 35:861-8. [DOI: 10.3892/or.2015.4460] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/10/2015] [Indexed: 11/06/2022] Open
|
625
|
Qin Y, Cheng C, Lu H, Wang Y. miR-4458 suppresses glycolysis and lactate production by directly targeting hexokinase2 in colon cancer cells. Biochem Biophys Res Commun 2015; 469:37-43. [PMID: 26607110 DOI: 10.1016/j.bbrc.2015.11.066] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 12/23/2022]
Abstract
miR-4458, a new tumor-suppressor, was reported to down-regulated in human hepatocellular carcinoma. The expression status, roles and inhibitory mechanisms of miR-4458 in other tumors still need to be clarified. The aim of this study is to investigate the effects of miR-4458 and to elucidate the potential mechanism in colon cancer cells. Using bioinformatic databases, we predicted that hexokinase2 (HK2), a rate-limiting enzyme in the glycolytic pathway, was a target of miR-4458, so the effects of miR-4458 on glycolysis and lactate production was assessed in colon cancer cells. We found that miR-4458 was down-regulated and HK2 was up-regulated in colon cancer cells. Overexpression of miR-4458 inhibited proliferation, glycolysis, and lactate production under both normoxic and hypoxic conditions. Luciferase activity assays showed that HK2 was a direct target of miR-4458. Moreover, knockdown of HK2 by specific RNAi also suppressed proliferation, glycolysis, and lactate production under both normoxic and hypoxic conditions. In conclusion, our findings suggested that miR-4458 inhibited the progression of colon cancer cells by inhibition of glycolysis and lactate production via directly targeting HK2 mRNA.
Collapse
Affiliation(s)
- Yaguang Qin
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, PR China
| | - Chuanyao Cheng
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, PR China
| | - Hong Lu
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, PR China.
| | - Yaqiu Wang
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, PR China
| |
Collapse
|
626
|
Metformin Induces Cell Cycle Arrest and Apoptosis in Drug-Resistant Leukemia Cells. LEUKEMIA RESEARCH AND TREATMENT 2015; 2015:516460. [PMID: 26688757 PMCID: PMC4673355 DOI: 10.1155/2015/516460] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/18/2015] [Accepted: 09/20/2015] [Indexed: 12/25/2022]
Abstract
Recent epidemiological studies indicate that the antidiabetic drug metformin has chemosensitizing and chemopreventive effects against carcinogenesis. Here, we demonstrate that metformin exerts varying degrees of antitumor activity against human leukemia cells, as reflected by differences in growth inhibition, apoptosis, and alterations to metabolic enzymes. In metformin-sensitive cells, autophagy was not induced but rather it blocked proliferation by means of arresting cells in the S and G2/M phases which was associated with the downregulation of cyclin A, cyclin B1, and cdc2, but not that of cyclin E. In 10E1-CEM cells that overexpress Bcl-2 and are drug-resistant, the effect of metformin on proliferation was more pronounced, also inducing the activation of the caspases 3/7 and hence apoptosis. In all sensitive cells, metformin decreased the Δψm and it modified the expression of enzymes involved in energy metabolism: PKCε (PKCepsilon) and PKCδ (PKCdelta). In sensitive cells, metformin altered PKCε and PKCδ expression leading to a predominance of PKCε over PKCδ which implies a more glycolytic state. The opposite occurs in the nonresponsive cells. In conclusion, we provide new insights into the activity of metformin as an antitumoral agent in leukemia cells that could be related to its capability to modulate energy metabolism.
Collapse
|
627
|
Hong SE, Jin HO, Kim HA, Seong MK, Kim EK, Ye SK, Choe TB, Lee JK, Kim JI, Park IC, Noh WC. Targeting HIF-1α is a prerequisite for cell sensitivity to dichloroacetate (DCA) and metformin. Biochem Biophys Res Commun 2015; 469:164-70. [PMID: 26616058 DOI: 10.1016/j.bbrc.2015.11.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 11/19/2015] [Indexed: 12/30/2022]
Abstract
Recently, targeting deregulated energy metabolism is an emerging strategy for cancer therapy. In the present study, combination of DCA and metformin markedly induced cell death, compared with each drug alone. Furthermore, the expression levels of glycolytic enzymes including HK2, LDHA and ENO1 were downregulated by two drugs. Interestingly, HIF-1α activation markedly suppressed DCA/metformin-induced cell death and recovered the expressions of glycolytic enzymes that were decreased by two drugs. Based on these findings, we propose that targeting HIF-1α is necessary for cancer metabolism targeted therapy.
Collapse
Affiliation(s)
- Sung-Eun Hong
- Division of Radiation Cancer Research, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Hyun-Ah Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Min-Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Eun-Kyu Kim
- Department of Surgery, Breast Cancer Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Republic of Korea
| | - Sang-Kyu Ye
- Department of Pharmacology, Seoul National University College of Medicine, 103 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea
| | - Tae-Boo Choe
- Department of Microbiological Engineering, Kon-Kuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Jong-Il Kim
- Department of Food Science & Technology, Seoul Women's University, 621 Hwarangro, Nowon-gu, Seoul 01797, Republic of Korea
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea.
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea.
| |
Collapse
|
628
|
Roh JL, Park JY, Kim EH, Jang HJ, Kwon M. Activation of mitochondrial oxidation by PDK2 inhibition reverses cisplatin resistance in head and neck cancer. Cancer Lett 2015; 371:20-9. [PMID: 26607904 DOI: 10.1016/j.canlet.2015.11.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 01/21/2023]
Abstract
Dichloroacetate (DCA), an orphan drug that promotes a shift from glycolysis to oxidative phosphorylation, has been repurposed for cancer therapy. The present study investigated whether DCA may overcome cisplatin resistance in head and neck cancer (HNC). Two cisplatin-resistant HNC cell lines (AMC-HN4R and -HN9R), their parental lines, and other human HNC lines were used. The effect of DCA, alone and in combination with cisplatin, was assessed by measuring cell cycle, viability, death, reactive oxygen species (ROS) production, mitochondrial membrane potential (ΔΨm), and protein expression in preclinical mouse tumor xenograft models. Increased glycolysis correlated with decreased sensitivity to cisplatin and was reduced by DCA. Cisplatin-resistant cells overexpressed pyruvate dehydrogenase kinase 2 (PDK2). DCA induced HNC cell death by decreasing ΔΨm and promoting mitochondrial ROS production. This effect was decreased by the antioxidant N-acetyl-l-cysteine or by inhibition of caspase-mediated apoptosis. Activation of mitochondrial glucose oxidation by DCA eventually activated downstream mitochondrial apoptotic signaling, leading to the death of chemoresistant cancer cells. Therefore, DCA significantly sensitized resistant HNC cells to cisplatin in vitro and in vivo. High glycolysis and PDK2 overexpression are closely linked to cisplatin resistance in HNC cells; the latter can be overcome by DCA.
Collapse
Affiliation(s)
- Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Jin Young Park
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye Jin Jang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minsu Kwon
- Department of Otolaryngology, Gyeongsang National University Hospital School of Medicine, Jinju, Republic of Korea
| |
Collapse
|
629
|
Lee YM, Lee G, Oh TI, Kim BM, Shim DW, Lee KH, Kim YJ, Lim BO, Lim JH. Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress. Int J Oncol 2015; 48:399-408. [PMID: 26573871 DOI: 10.3892/ijo.2015.3243] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/20/2015] [Indexed: 11/06/2022] Open
Abstract
Recent studies have shown anticancer activity of apigenin by suppressing glucose transporter 1 (GLUT1) expression in cultured cancer cells; however, it is not clear whether apigenin can suppress glucose metabolism in lung cancer cells or sensitize them to inhibition of glutamine utilization-mediated apoptosis through metabolic and oxidative stress. We show that apigenin significantly decreases GLUT1 expression in mice. Furthermore, we demonstrate that apigenin induces growth retardation and apoptosis through metabolic and oxidative stress caused by suppression of glucose utilization in lung cancer cells. The underlying mechanisms were defined that the anticancer effects of apigenin were reversed by ectopic GLUT1 overexpression and galactose supplementation, through activation of pentose phosphate pathway-mediated NADPH generation. Importantly, we showed that severe metabolic stress using a glutaminase inhibitor, compound 968, was involved in the mechanism of sensitization by apigenin. Taken together, the combination of apigenin with inhibitors of glutamine metabolism may provide a promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Yoon-Mi Lee
- Department of Food Bioscience, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Gibok Lee
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Taek-In Oh
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Byeong Mo Kim
- Severance Integrative Research Institute for Cerebral and Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, Seodaemun-gu, Seoul 120-752, Republic of Korea
| | - Do-Wan Shim
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Kwang-Ho Lee
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Young Jun Kim
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Beong Ou Lim
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| | - Ji-Hong Lim
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungbuk, Chungju 380-701, Republic of Korea
| |
Collapse
|
630
|
Amelio I, Antonov AA, Catani MV, Massoud R, Bernassola F, Knight RA, Melino G, Rufini A. TAp73 promotes anabolism. Oncotarget 2015; 5:12820-934. [PMID: 25514460 PMCID: PMC4350352 DOI: 10.18632/oncotarget.2667] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 12/18/2022] Open
Abstract
Metabolic adaptation has emerged as a hallmark of cancer and a promising therapeutic target, as rapidly proliferating cancer cells adapt their metabolism increasing nutrient uptake and reorganizing metabolic fluxes to support biosynthesis. The transcription factor p73 belongs to the p53-family and regulates tumorigenesis via its two N-terminal isoforms, with (TAp73) or without (ΔNp73) a transactivation domain. TAp73 acts as tumor suppressor, at least partially through induction of cell cycle arrest and apoptosis and through regulation of genomic stability. Here, we sought to investigate whether TAp73 also affects metabolic profiling of cancer cells. Using high throughput metabolomics, we unveil a thorough and unexpected role for TAp73 in promoting Warburg effect and cellular metabolism. TAp73-expressing cells show increased rate of glycolysis, higher amino acid uptake and increased levels and biosynthesis of acetyl-CoA. Moreover, we report an extensive TAp73-mediated upregulation of several anabolic pathways including polyamine and synthesis of membrane phospholipids. TAp73 expression also increases cellular methyl-donor S-adenosylmethionine (SAM), possibly influencing methylation and epigenetics, and promotes arginine metabolism, suggestive of a role in extracellular matrix (ECM) modeling. In summary, our data indicate that TAp73 regulates multiple metabolic pathways that impinge on numerous cellular functions, but that, overall, converge to sustain cell growth and proliferation.
Collapse
Affiliation(s)
- Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Alexey A Antonov
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Maria Valeria Catani
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Renato Massoud
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Francesca Bernassola
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Richard A Knight
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK. Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy. Molecular Pharmacology Laboratory, Technological University, St-Petersburg, Russia
| | - Alessandro Rufini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK. Department of Cancer Studies, Cancer Research UK, Leicester Centre, University of Leicester, Leicester, LE1 7RH, UK
| |
Collapse
|
631
|
Diedrich J, Gusky HC, Podgorski I. Adipose tissue dysfunction and its effects on tumor metabolism. Horm Mol Biol Clin Investig 2015; 21:17-41. [PMID: 25781550 DOI: 10.1515/hmbci-2014-0045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/14/2015] [Indexed: 12/12/2022]
Abstract
Growing by an alarming rate in the Western world, obesity has become a condition associated with a multitude of diseases such as diabetes, metabolic syndrome and various cancers. Generally viewed as an abnormal accumulation of hypertrophied adipocytes, obesity is also a poor prognostic factor for recurrence and chemoresistance in cancer patients. With more than two-thirds of the adult population in the United States considered clinically overweight or obese, it is critical that the relationship between obesity and cancer is further emphasized and elucidated. Adipocytes are highly metabolically active cells, which, through release of adipokines and cytokines and activation of endocrine and paracrine pathways, affect processes in neighboring and distant cells, altering their normal homeostasis. This work will examine specifically how adipocyte-derived factors regulate the cellular metabolism of malignant cells within the tumor niche. Briefly, tumor cells undergo metabolic pressure towards a more glycolytic and hypoxic state through a variety of metabolic regulators and signaling pathways, i.e., phosphoinositol-3 kinase (PI3K), hypoxia-inducible factor-1 alpha (HIF-1α), and c-MYC signaling. Enhanced glycolysis and high lactate production are hallmarks of tumor progression largely because of a process known as the Warburg effect. Herein, we review the latest literature pertaining to the body of work on the interactions between adipose and tumor cells, and underlining the changes in cancer cell metabolism that have been targeted by the currently available treatments.
Collapse
|
632
|
Zhang W, Zhang SL, Hu X, Tam KY. Targeting Tumor Metabolism for Cancer Treatment: Is Pyruvate Dehydrogenase Kinases (PDKs) a Viable Anticancer Target? Int J Biol Sci 2015; 11:1390-400. [PMID: 26681918 PMCID: PMC4671996 DOI: 10.7150/ijbs.13325] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/07/2015] [Indexed: 01/07/2023] Open
Abstract
Cancer remains a lethal threat to global lives. Development of novel anticancer therapeutics is still a challenge to scientists in the field of biomedicine. In cancer cells, the metabolic features are significantly different from those of normal ones, which are hallmarks of several malignancies. Recent studies brought atypical cellular metabolism, such as aerobic glycolysis or the Warburg effect, into the scientific limelight. Targeting these altered metabolic pathways in cancer cells presents a promising therapeutic strategy. Pyruvate dehydrogenase kinases (PDKs), key enzymes in the pathway of glucose metabolism, could inactivate the pyruvate dehydrogenase complex (PDC) by phosphorylating it and preserving the substrates pyruvate, lactate and alanine for gluconeogenesis. Overexpression of PDKs could block the oxidative decarboxylation of pyruvate to satisfy high oxygen demand in cancer cells, while inhibition of PDKs could upregulate the activity of PDC and rectify the balance between the demand and supply of oxygen, which could lead to cancer cell death. Thus, inhibitors targeting PDKs represent a promising strategy for cancer treatment by acting on glycolytic tumors while showing minimal side effects on the oxidative healthy organs. This review considers the role of PDKs as regulator of PDC that catalyzes the oxidative decarboxylation of pyruvate in mitochondrion. It is concluded that PDKs are solid therapeutic targets. Inhibition of PDKs could be an attractive therapeutic approach for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- Wen Zhang
- Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Shao-Lin Zhang
- Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaohui Hu
- Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China
| | - Kin Yip Tam
- Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
633
|
Arambula JF, McCall R, Sidoran KJ, Magda D, Mitchell NA, Bielawski CW, Lynch VM, Sessler JL, Arumugam K. Targeting Antioxidant Pathways with Ferrocenylated N-Heterocyclic Carbene Supported Gold(I) Complexes in A549 Lung Cancer Cells. Chem Sci 2015; 7:1245-1256. [PMID: 26918111 PMCID: PMC4762604 DOI: 10.1039/c5sc03519h] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Ferrocenylated-Au(i) carbenes were designed, synthesized, and studied for their ability to generate reactive oxygen species and target antioxidant pathways via multiple mechanisms.
Ferrocene containing N-heterocyclic carbene (NHC) ligated gold(i) complexes of the type [Au(NHC)2]+ were prepared and found to be capable of regulating the formation of reactive oxygen species (ROS) via multiple mechanisms. Single crystal X-ray analysis of bis(1-(ferrocenylmethyl)-3-mesitylimidazol-2-ylidene)-gold(i) chloride (5) and bis(1,3-di(ferrocenylmethyl)imidazol-2-ylidene)-gold(i) chloride (6) revealed a quasi-linear geometry around the gold(i) centers (i.e., the C–Au–C bond angle were measured to be ∼177° and all the Au–Ccarbene bonds distances were in the range of 2.00 (7)–2.03 (1) Å). A series of cell studies indicated that cell proliferation inhibition and ROS generation were directly proportional to the amount of ferrocene contained within the [Au(NHC)2]+ complexes (IC50 of 6 < 5 < bis(1-benzyl-3-mesitylimidazol-2-ylidene)-gold(i) chloride (4)). Complexes 4–6 were also confirmed to inhibit thioredoxin reductase as inferred from lipoate reduction assays and increased chelatable intracellular zinc concentrations. RNA microarray gene expression assays revealed that 6 induces endoplasmic reticulum stress response pathways as a result of ROS increase.
Collapse
Affiliation(s)
- J F Arambula
- Department of Chemistry, Georgia Southern University, Statesboro, Georgia, 30460, USA
| | - R McCall
- Department of Chemistry, Georgia Southern University, Statesboro, Georgia, 30460, USA
| | - K J Sidoran
- Department of Chemistry, 3261 West State Road, St. Bonaventure University, New York, 14778
| | - D Magda
- Lumiphore, Inc., Berkeley, California, 94710, USA
| | - N A Mitchell
- Department of Health Sciences, Gettysburg College, Gettysburg, PA 17325-1400
| | - C W Bielawski
- Center for Multidimensional Carbon Materials (CMCM), Institute for Basic Science (IBS), Ulsan 689-798, Republic of Korea; Department of Chemistry and Department of Energy Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Republic of Korea
| | - V M Lynch
- Department of Chemistry, University of Texas at Austin, Austin, Texas, 78712, USA
| | - J L Sessler
- Department of Chemistry, University of Texas at Austin, Austin, Texas, 78712, USA
| | - K Arumugam
- Department of Chemistry, Wright State University, 3640 Colonel Glenn Hwy, Dayton, Ohio, 45435, USA
| |
Collapse
|
634
|
Shibuya K, Okada M, Suzuki S, Seino M, Seino S, Takeda H, Kitanaka C. Targeting the facilitative glucose transporter GLUT1 inhibits the self-renewal and tumor-initiating capacity of cancer stem cells. Oncotarget 2015; 6:651-61. [PMID: 25528771 PMCID: PMC4359246 DOI: 10.18632/oncotarget.2892] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022] Open
Abstract
Increased glucose metabolism is now recognized as an emerging hallmark of cancer. Recent studies have shown that glucose metabolism is even more active in cancer stem cells (CSCs), a rare population of cancer cells with the capacity to self-renew and initiate tumors, and that CSCs are dependent on glycolysis for their survival/growth. However, the role of glucose metabolism in the control of their self-renewal and tumor-initiating capacity per se still remains obscure. Moreover, much remains unknown as to which of the numerous molecules involved in the glucose metabolism is suitable as a target to control CSCs. Here we demonstrate that the facilitative glucose transporter GLUT1 is essential for the maintenance of pancreatic, ovarian, and glioblastoma CSCs. Notably, we found that WZB117, a specific GLUT1 inhibitor, could inhibit the self-renewal and tumor-initiating capacity of the CSCs without compromising their proliferative potential in vitro. In vivo, systemic WZB117 administration inhibited tumor initiation after implantation of CSCs without causing significant adverse events in host animals. Our findings indicate GLUT1-dependent glucose metabolism has a pivotal role not only in the growth and survival of CSCs but also in the maintenance of their stemness and suggest GLUT1 as a promising target for CSC-directed cancer therapy.
Collapse
Affiliation(s)
- Keita Shibuya
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.,Oncology Research Center, Research Institute for Advanced Molecular Epidemiology, Yamagata University, Yamagata, Japan.,Global COE program for Medical Sciences, Japan Society for Promotion of Science, Tokyo, Japan
| | - Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.,Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Manabu Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.,Department of Obstetrics and Gynecology, Yamagata University School of Medicine, Yamagata, Japan
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.,Oncology Research Center, Research Institute for Advanced Molecular Epidemiology, Yamagata University, Yamagata, Japan.,Global COE program for Medical Sciences, Japan Society for Promotion of Science, Tokyo, Japan.,Research Institute for Promotion of Medical Sciences, Yamagata University School of Medicine, Yamagata, Japan
| | - Hiroyuki Takeda
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.,Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan.Oncology Research Center, Research Institute for Advanced Molecular Epidemiology, Yamagata University, Yamagata, Japan.Global COE program for Medical Sciences, Japan Society for Promotion of Science, Tokyo, Japan.Research Institute for Promotion of Medical Sciences, Yamagata University School of Medicine, Yamagata, Japan
| |
Collapse
|
635
|
Honndorf VS, Schmidt H, Wehrl HF, Wiehr S, Ehrlichmann W, Quintanilla-Martinez L, Barjat H, Ricketts SA, Pichler BJ. Quantitative correlation at the molecular level of tumor response to docetaxel by multimodal diffusion-weighted magnetic resonance imaging and [¹⁸F]FDG/[¹⁸F]FLT positron emission tomography. Mol Imaging 2015; 13. [PMID: 25430886 DOI: 10.2310/7290.2014.00045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
We aimed to quantitatively characterize the treatment effects of docetaxel in the HCT116 xenograft mouse model, applying diffusion-weighted magnetic resonance imaging (MRI) and positron emission tomography (PET) using 2-deoxy-2-[¹⁸F]fluoro-d-glucose ([¹⁸F]FDG) and 3'-deoxy-3'-[¹⁸F]-fluorothymidine ([¹⁸F]FLT). Mice were imaged at four time points over 8 days. Docetaxel (15 mg/kg) was administered after a baseline scan. Voxel-wise scatterplots of PET and apparent diffusion coefficient (ADC) data of tumor volumes were evaluated with a threshold cluster analysis and compared to histology (GLUT1, GLUT3, Ki67, activated caspase 3a). Compared to the extensive tumor growth observed in the vehicle-treated group (from 0.32 ± 0.21 cm³ to 0.69 ± 0.40 cm³), the administration of docetaxel led to tumor growth stasis (from 0.32 ± 0.20 cm³ to 0.45 ± 0.23 cm³). The [¹⁸F]FDG/ADC cluster analysis and the evaluation of peak histogram values revealed a significant treatment effect matching histology as opposed to [¹⁸F]FLT/ADC. [¹⁸F]FLT uptake and the Ki67 index were not in good agreement. Our voxel-based cluster analysis uncovered treatment effects not seen in the separate inspection of PET and MRI data and may be used as an independent analysis tool. [¹⁸F]FLT/ADC cluster analysis could still point out the treatment effect; however, [¹⁸F]FDG/ADC reflected the histology findings in higher agreement.
Collapse
|
636
|
Lactate dehydrogenase-A inhibition induces human glioblastoma multiforme stem cell differentiation and death. Sci Rep 2015; 5:15556. [PMID: 26494310 PMCID: PMC4616042 DOI: 10.1038/srep15556] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
Therapies that target the signal transduction and metabolic pathways of cancer stem cells (CSCs) are innovative strategies to effectively reduce the recurrence and significantly improve the outcome of glioblastoma multiforme (GBM). CSCs exhibit an increased rate of glycolysis, thus rendering them intrinsically more sensitive to prospective therapeutic strategies based on the inhibition of the glycolytic pathway. The enzyme lactate dehydrogenase-A (LDH-A), which catalyses the interconversion of pyruvate and lactate, is up-regulated in human cancers, including GBM. Although several papers have explored the benefits of targeting cancer metabolism in GBM, the effects of direct LDH-A inhibition in glial tumours have not yet been investigated, particularly in the stem cell subpopulation. Here, two representative LDH-A inhibitors (NHI-1 and NHI-2) were studied in GBM-derived CSCs and compared to differentiated tumour cells. LDH-A inhibition was particularly effective in CSCs isolated from different GBM cell lines, where the two compounds blocked CSC formation and elicited long-lasting effects by triggering both apoptosis and cellular differentiation. These data demonstrate that GBM, particularly the stem cell subpopulation, is sensitive to glycolytic inhibition and shed light on the therapeutic potential of LDH-A inhibitors in this tumour type.
Collapse
|
637
|
Kim SH, Lee JE, Sharker SM, Jeong JH, In I, Park SY. In Vitro and In Vivo Tumor Targeted Photothermal Cancer Therapy Using Functionalized Graphene Nanoparticles. Biomacromolecules 2015; 16:3519-29. [PMID: 26451914 DOI: 10.1021/acs.biomac.5b00944] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the tremendous progress that photothermal therapy (PTT) has recently achieved, it still has a long way to go to gain the effective targeted photothermal ablation of tumor cells. Driven by this need, we describe a new class of targeted photothermal therapeutic agents for cancer cells with pH responsive bioimaging using near-infrared dye (NIR) IR825, conjugated poly(ethylene glycol)-g-poly(dimethylaminoethyl methacrylate) (PEG-g-PDMA, PgP), and hyaluronic acid (HA) anchored reduced graphene oxide (rGO) hybrid nanoparticles. The obtained rGO nanoparticles (PgP/HA-rGO) showed pH-dependent fluorescence emission and excellent near-infrared (NIR) irradiation of cancer cells targeted in vitro to provide cytotoxicity. Using intravenously administered PTT agents, the time-dependent in vivo tumor target accumulation was exactly defined, presenting eminent photothermal conversion at 4 and 8 h post-injection, which was demonstrated from the ex vivo biodistribution of tumors. These tumor environment responsive hybrid nanoparticles generated photothermal heat, which caused dominant suppression of tumor growth. The histopathological studies obtained by H&E staining demonstrated complete healing from malignant tumor. In an area of limited successes in cancer therapy, our translation will pave the road to design stimulus environment responsive targeted PTT agents for the safe eradication of devastating cancer.
Collapse
Affiliation(s)
| | - Jung Eun Lee
- School of Pharmacy, Sungkyunkwan University , 300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | - Shazid Md Sharker
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 305-701, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University , 300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do 440-746, Republic of Korea
| | | | | |
Collapse
|
638
|
Mandai M, Amano Y, Yamaguchi K, Matsumura N, Baba T, Konishi I. Ovarian clear cell carcinoma meets metabolism; HNF-1β confers survival benefits through the Warburg effect and ROS reduction. Oncotarget 2015; 6:30704-14. [PMID: 26375553 PMCID: PMC4741562 DOI: 10.18632/oncotarget.5228] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) constitutes one of the subtypes of ovarian cancers, but it has unique clinical, histological and biological characteristics, one of which is chemo-resistance. It is also known to develop from endometriotic cyst, a benign ovarian tumor, at relatively high frequency. Recently, it is becoming well known that most of OCCCs express HNF1β, a transcription factor, which is closely associated with the development of liver, pancreas and kidney, as well as occurrence of familial forms of type 2 diabetes. Expression of HNF1β is now regarded as a hallmark of this tumor. Nevertheless, exact biological function of this gene in OCCC has not been clarified. We have shown in previous studies that microenvironment in endometriotic cysts contains severe oxidative stress and OCCC develops under such stressful environment as stress-resistant tumor, which may lead to chemo-resistance. We also showed that increased expression of HNF1β facilitates glucose uptake and glycolysis, which is known as Warburg effect. In the previous issue of this journal, by using comprehensive metabolome analysis, we report that HNF1β actually reduces and protects themselves from internal oxidative stress by dramatically changing cellular metabolism. In this article, we review the relevance and significance of cancer-specific metabolism and how they are associated with biological characteristics of OCCC via expression of HNF1β, along with future clinical implications of targeting cancer-specific metabolism.
Collapse
Affiliation(s)
- Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Obstetrics and Gynecology, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasuaki Amano
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
639
|
Abstract
Thermally responsive nanogel drug delivery systems (TRNDDS) have been widely investigated as a new strategy for active targeting tumor therapy, as these can accumulate on the tumor site and/or release the payload at the desired site by structure changes rapidly once stimulated by temperature changes. In this review, we discuss the evolution of TRNDDS and future perspectives for antitumor drug and gene delivery. With further understanding of the specificity of tumor site at the cellular and molecular level, in parallel with the development of nanomaterial design and preparation, TRNDDS show great potential for tumor targeting therapy.
Collapse
|
640
|
Guan DG, Chen HM, Liao SF, Zhao TZ. Combination of temozolomide and Taxol exerts a synergistic inhibitory effect on Taxol‑resistant glioma cells via inhibition of glucose metabolism. Mol Med Rep 2015; 12:7705-11. [PMID: 26459853 DOI: 10.3892/mmr.2015.4405] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/11/2015] [Indexed: 11/06/2022] Open
Abstract
Malignant gliomas, which comprise the most common type of primary malignant brain tumor, are associated with a poor prognosis and quality of life. Paclitaxel (Taxol) and temozolomide (TMZ) are Food and Drug Administration‑approved anticancer agents, which are known to have therapeutic applications in various malignancies. However, similar to other chemotherapeutic agents, the development of resistance to TMZ and Taxol is common. The aim of the present study was to investigate the regulation of glucose metabolism by TMZ and Taxol in glioma cells. The results demonstrated that glioma cells exhibit decreased glucose uptake and lactate production in response to treatment with TMZ; however, glucose metabolism was increased in response to Taxol treatment. Following analysis of TMZ‑ and Taxol‑resistant cell lines, it was reported that glucose metabolism was decreased in the TMZ‑resistant cells, but was increased in the Taxol‑resistant cells. Notably, a combination of TMZ and Taxol exerted synergistic inhibitory effects on Taxol‑resistant glioma cells. However, the synergistic phenotype was not observed following treatment with a combination of 5‑fluorouracil and Taxol. Furthermore, restoration of glucose metabolism by overexpression of glucose transporter 1 in Taxol‑resistant cells resulted in regained resistance to Taxol. Therefore, the present study proposes a novel mechanism accounting for the synergistic effects of Taxol and TMZ co‑treatment, which may contribute to the development of therapeutic strategies for overcoming chemoresistance in patients with cancer.
Collapse
Affiliation(s)
- Ding-Guo Guan
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Han-Min Chen
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Sheng-Fang Liao
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Tian-Zhi Zhao
- Department of Neurosurgery, Affiliated Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shanxi 710038, P.R. China
| |
Collapse
|
641
|
Mitra K, Gautam S, Kondaiah P, Chakravarty AR. Thecis-Diammineplatinum(II) Complex of Curcumin: A Dual Action DNA Crosslinking and Photochemotherapeutic Agent. Angew Chem Int Ed Engl 2015; 54:13989-93. [DOI: 10.1002/anie.201507281] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 08/19/2015] [Indexed: 01/17/2023]
|
642
|
Baiu DC, Artz NS, McElreath MR, Menapace BD, Hernando D, Reeder SB, Grüttner C, Otto M. High specificity targeting and detection of human neuroblastoma using multifunctional anti-GD2 iron-oxide nanoparticles. Nanomedicine (Lond) 2015; 10:2973-2988. [PMID: 26420448 DOI: 10.2217/nnm.15.138] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM To develop biocompatible, tumor-specific multifunctional iron-oxide nanoconstructs targeting neuroblastoma, an aggressive pediatric malignancy. MATERIALS & METHODS Clinical-grade humanized monoclonal antibody (hu14.18K322A), designed to target GD2 antigen on neuroblastoma with reduced nonspecific immune interactions, was conjugated to hydroxyethyl starch-coated iron-oxide nanoparticles. Targeting capability in vitro and in vivo was assessed by immunofluorescence, electron microscopy, analytical spectrophotometry, histochemistry and magnetic resonance R2* relaxometry. RESULTS The biocompatible nanoconstructs demonstrated high tumor specificity in vitro and in vivo, and low background uptake in a mouse flank xenograft model. Specific accumulation in tumors enabled particle visualization and quantification by magnetic resonance R2* mapping. CONCLUSION Our findings support the further development toward clinical application of this anti-GD2 iron-oxide nanoconstruct as diagnostic and therapeutic scaffold for neuroblastoma and potentially other GD2-positive malignancies.
Collapse
Affiliation(s)
- Dana C Baiu
- Department of Pediatrics, Division of Pediatric Hematology, Oncology & Bone Marrow Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nathan S Artz
- Department of Radiology, Medical Physics, Biomedical Engineering, Medicine & Emergency Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Meghan R McElreath
- Department of Pediatrics, Division of Pediatric Hematology, Oncology & Bone Marrow Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bryan D Menapace
- Department of Pediatrics, Division of Pediatric Hematology, Oncology & Bone Marrow Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Diego Hernando
- Department of Radiology, Medical Physics, Biomedical Engineering, Medicine & Emergency Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Scott B Reeder
- Department of Radiology, Medical Physics, Biomedical Engineering, Medicine & Emergency Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Mario Otto
- Department of Pediatrics, Division of Pediatric Hematology, Oncology & Bone Marrow Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
643
|
Mitra K, Gautam S, Kondaiah P, Chakravarty AR. The
cis
‐Diammineplatinum(II) Complex of Curcumin: A Dual Action DNA Crosslinking and Photochemotherapeutic Agent. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201507281] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Koushambi Mitra
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka (India)
| | - Srishti Gautam
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, Karnataka (India)
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, Karnataka (India)
| | - Akhil R. Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka (India)
| |
Collapse
|
644
|
Hansen JS, Elbing K, Thompson JR, Malmstadt N, Lindkvist-Petersson K. Glucose transport machinery reconstituted in cell models. Chem Commun (Camb) 2015; 51:2316-9. [PMID: 25562394 DOI: 10.1039/c4cc08838g] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Here we demonstrate the production of a functioning cell model by formation of giant vesicles reconstituted with the GLUT1 glucose transporter and a glucose oxidase and hydrogen peroxidase linked fluorescent reporter internally. Hence, a simplified artificial cell is formed that is able to take up glucose and process it.
Collapse
Affiliation(s)
- Jesper S Hansen
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Box 117, SE-22100, Lund, Sweden.
| | | | | | | | | |
Collapse
|
645
|
von Roemeling CA, Copland JA. Targeting lipid metabolism for the treatment of anaplastic thyroid carcinoma. Expert Opin Ther Targets 2015; 20:159-66. [PMID: 26414044 DOI: 10.1517/14728222.2016.1086341] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Anaplastic thyroid carcinoma (ATC) is the rarest subtype of thyroid cancer; however, it disproportionately accounts for a large percentage of all thyroid cancer-related deaths and is considered one of the most lethal solid tumors in humans, having a median survival of only a few months upon diagnosis. Although a variety of treatment options are available including surgery, radiation and targeted therapies, response rates are low, due in part to the drug-resistant nature of this disease; therefore, new avenues for therapeutic intervention are surely needed. Recent investigation into the metabolic profile of ATC has revealed a tumor-specific dependency for increased de novo lipogenesis, offering new insight into the molecular mechanisms that govern disease initiation and progression. AREAS COVERED Herein we summarize known oncogenic signaling pathways and current therapeutic strategies for the treatment of ATC. We further discuss the unique expression pattern of lipid metabolism constituents in this disease. Additionally, the current literature correlating aberrant lipogenesis with carcinogenesis is reviewed, and the implications of targeting this pathway as an innovative approach for treating ATC and other malignancies are discussed. As stearoyl-CoA desaturase (SCD) is the most differentially expressed constituent of lipid metabolism in ATC, an additional focus on this enzyme as a novel therapeutic target is applied. EXPERT OPINION This section is used to summarize the current research efforts underway in defining the role of lipid metabolism specifically in thyroid carcinoma. Included is a brief summary of lipid metabolism factors for which inhibitors have been generated and are under current investigation as anti-cancer agents. Finally, research limitations regarding the use of these inhibitors against components of this pathway are discussed.
Collapse
Affiliation(s)
| | - John A Copland
- b 2 Mayo Clinic Jacksonville, The Department of Cancer Biology , Jacksonville, FL 32224, USA ;
| |
Collapse
|
646
|
Identification of the determinants of 2-deoxyglucose sensitivity in cancer cells by shRNA library screening. Biochem Biophys Res Commun 2015; 467:121-7. [PMID: 26403972 DOI: 10.1016/j.bbrc.2015.09.106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/19/2015] [Indexed: 11/20/2022]
Abstract
Combining glycolytic inhibition with other anti-cancer therapies is a potential approach to treating cancer. In this context, we attempted to identify genes that determine sensitivity to 2-deoxyglucose (2DG), a glycolytic inhibitor, in cancer cells using pooled shRNA libraries targeting ∼15,000 genes. The screen revealed that COPB1 and ARCN1, which are essential in retrograde transport, as determinants of sensitivity to 2DG: silencing of COPB1 or ARCN1 expression sensitized cells to 2DG toxicity. To address the mechanism of potentiation of 2DG toxicity by inhibition of COPI-mediated transport, we focused on the role of lipolysis as an alternate source of energy upon inhibition of glycolysis. In the process of lipolysis, COPI-mediated transport is required for localization to lipid droplets of adipose triglyceride lipase (ATGL), a key enzyme that produces fatty acids from triacylglycerol as a substrate for β-oxidation. The ATGL inhibitor atglistatin potentiated 2DG toxicity, consistent with a model in which a defect in COPI-mediated transport of ATGL to lipid droplets inhibits energy supply, thereby sensitizing cells to glycolytic inhibition. Collectively, our data demonstrated that a defect in COPI-mediated transport or pharmacological inhibition of ATGL potentiates 2DG toxicity in cancer cells, possibly due to a reduction in the energy supply.
Collapse
|
647
|
Xintaropoulou C, Ward C, Wise A, Marston H, Turnbull A, Langdon SP. A comparative analysis of inhibitors of the glycolysis pathway in breast and ovarian cancer cell line models. Oncotarget 2015; 6:25677-95. [PMID: 26259240 PMCID: PMC4694858 DOI: 10.18632/oncotarget.4499] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/29/2015] [Indexed: 02/04/2023] Open
Abstract
Many cancer cells rely on aerobic glycolysis for energy production and targeting of this pathway is a potential strategy to inhibit cancer cell growth. In this study, inhibition of five glycolysis pathway molecules (GLUT1, HKII, PFKFB3, PDHK1 and LDH) using 9 inhibitors (Phloretin, Quercetin, STF31, WZB117, 3PO, 3-bromopyruvate, Dichloroacetate, Oxamic acid, NHI-1) was investigated in panels of breast and ovarian cancer cell line models. All compounds tested blocked glycolysis as indicated by increased extracellular glucose and decreased lactate production and also increased apoptosis. Sensitivity to several inhibitors correlated with the proliferation rate of the cell lines. Seven compounds had IC50 values that were associated with each other consistent with a shared mechanism of action. A synergistic interaction was revealed between STF31 and Oxamic acid when combined with the antidiabetic drug metformin. Sensitivity to glycolysis inhibition was also examined under a range of O2 levels (21% O2, 7% O2, 2% O2 and 0.5% O2) and greater resistance to the inhibitors was found at low oxygen conditions (7% O2, 2% O2 and 0.5% O2) relative to 21% O2 conditions. These results indicate growth of breast and ovarian cancer cell lines is dependent on all the targets examined in the glycolytic pathway with increased sensitivity to the inhibitors under normoxic conditions.
Collapse
Affiliation(s)
- Chrysi Xintaropoulou
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Carol Ward
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Alan Wise
- IOMET Pharma, Nine, Edinburgh BioQuarter, Edinburgh, EH16 4UX, UK
| | - Hugh Marston
- IOMET Pharma, Nine, Edinburgh BioQuarter, Edinburgh, EH16 4UX, UK
- Current Address: Eli Lilly Research and Development, Windlesham, Surrey, GU20 6PH, UK
| | - Arran Turnbull
- Breakthrough Breast Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Simon P. Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| |
Collapse
|
648
|
Chen L, Cui H. Targeting Glutamine Induces Apoptosis: A Cancer Therapy Approach. Int J Mol Sci 2015; 16:22830-55. [PMID: 26402672 PMCID: PMC4613338 DOI: 10.3390/ijms160922830] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Glutamine metabolism has been proved to be dysregulated in many cancer cells, and is essential for proliferation of most cancer cells, which makes glutamine an appealing target for cancer therapy. In order to be well used by cells, glutamine must be transported to cells by specific transporters and converted to glutamate by glutaminase. There are currently several drugs that target glutaminase under development or clinical trials. Also, glutamine metabolism restriction has been proved to be effective in inhibiting tumor growth both in vivo and vitro through inducing apoptosis, growth arrest and/or autophagy. Here, we review recent researches about glutamine metabolism in cancer, and cell death induced by targeting glutamine, and their potential roles in cancer therapy.
Collapse
Affiliation(s)
- Lian Chen
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Ya'an 625014, China.
| | - Hengmin Cui
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
649
|
Valvona CJ, Fillmore HL, Nunn PB, Pilkington GJ. The Regulation and Function of Lactate Dehydrogenase A: Therapeutic Potential in Brain Tumor. Brain Pathol 2015; 26:3-17. [PMID: 26269128 PMCID: PMC8029296 DOI: 10.1111/bpa.12299] [Citation(s) in RCA: 385] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/05/2015] [Indexed: 12/14/2022] Open
Abstract
There are over 120 types of brain tumor and approximately 45% of primary brain tumors are gliomas, of which glioblastoma multiforme (GBM) is the most common and aggressive with a median survival rate of 14 months. Despite progress in our knowledge, current therapies are unable to effectively combat primary brain tumors and patient survival remains poor. Tumor metabolism is important to consider in therapeutic approaches and is the focus of numerous research investigations. Lactate dehydrogenase A (LDHA) is a cytosolic enzyme, predominantly involved in anaerobic and aerobic glycolysis (the Warburg effect); however, it has multiple additional functions in non‐neoplastic and neoplastic tissues, which are not commonly known or discussed. This review summarizes what is currently known about the function of LDHA and identifies areas that would benefit from further exploration. The current knowledge of the role of LDHA in the brain and its potential as a therapeutic target for brain tumors will also be highlighted. The Warburg effect appears to be universal in tumors, including primary brain tumors, and LDHA (because of its involvement with this process) has been identified as a potential therapeutic target. Currently, there are, however, no suitable LDHA inhibitors available for tumor therapies in the clinic.
Collapse
Affiliation(s)
- Cara J Valvona
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Helen L Fillmore
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Peter B Nunn
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Geoffrey J Pilkington
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| |
Collapse
|
650
|
Friesen DE, Baracos VE, Tuszynski JA. Modeling the energetic cost of cancer as a result of altered energy metabolism: implications for cachexia. Theor Biol Med Model 2015; 12:17. [PMID: 26370269 PMCID: PMC4570294 DOI: 10.1186/s12976-015-0015-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/01/2015] [Indexed: 01/06/2023] Open
Abstract
Background Cachexia affects most patients with incurable cancer. We hypothesize that in metastatic cancer the mass of the tumor as well as its level of anaerobic energy metabolism play a critical role in describing its energetic cost, which results in elevated resting energy expenditure and glucose utilization, leading to cachexia. Prior models of cancer cachexia may have underestimated the specific energetic cost of cancer as they have not taken the range of tumor mass and anaerobic energy metabolism fully into account. Methods We therefore modelled the energetic cost of cancer as a function of the percentage of energy the cancer produces anaerobically, based on resting energy expenditure, glucose turnover, glucose recycling, and oxygen consumption in cancer patients found in previous studies. Results Data from two clinical studies where tumor burden was estimated and resting energy expenditure or oxygen consumption were measured lead to a broad range of estimates of tumor cost from 190 to 470 kcal/kg tumor/day. These values will vary based of the percentage of energy the cancer produces anaerobically (from 0 to 100 %), which in and of itself can alter the cost over a 2 to 3-fold range. In addition to the tumor cost/kg and the degree of anaerobic metabolism, the impact on a given individual patient will depend on tumor burden, which can exceed 1 kg in advanced metastatic disease. Considering these dimensions of tumor cost we are able to produce a 2-dimensional map of potential values, with an overall range of 100–1400 kcal/day. Conclusions Quantifying the energetic cost of cancer may benefit an understanding of the tumor’s causation of cachexia. Our estimates of the range of tumor cost include values that are higher than prior estimates and suggest that in metastatic disease the tumor cost could be expected to eclipse attempts to stabilize energy balance through nutrition support or by drug therapies. Tumor mass and the percentage of anaerobic metabolism in the tumor contribute to the cost of the tumor on the body and potentially lead directly to negative energy balance and increased muscle wasting. Electronic supplementary material The online version of this article (doi:10.1186/s12976-015-0015-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Douglas E Friesen
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 1Z2, Canada.
| | - Vickie E Baracos
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 1Z2, Canada.
| | - Jack A Tuszynski
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 1Z2, Canada. .,Department of Physics, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|