251
|
Byeon HK, Ku M, Yang J. Beyond EGFR inhibition: multilateral combat strategies to stop the progression of head and neck cancer. Exp Mol Med 2019; 51:1-14. [PMID: 30700700 PMCID: PMC6353966 DOI: 10.1038/s12276-018-0202-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/03/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) overexpression is common in head and neck squamous cell carcinoma. Targeted therapy specifically directed towards EGFR has been an area of keen interest in head and neck cancer research, as EGFR is potentially an integration point for convergent signaling. Despite the latest advancements in cancer diagnostics and therapeutics against EGFR, the survival rates of patients with advanced head and neck cancer remain disappointing due to anti-EGFR resistance. This review article will discuss recent multilateral efforts to discover and validate actionable strategies that involve signaling pathways in heterogenous head and neck cancer and to overcome anti-EGFR resistance in the era of precision medicine. Particularly, this review will discuss in detail the issue of cancer metabolism, which has recently emerged as a novel mechanism by which head and neck cancer may be successfully controlled according to different perspectives. South Korean researchers propose novel combination strategies for overcoming drug resistance and halting the progression of head and neck cancer (HNC). Although high levels of epidermal growth factor receptor (EGFR) protein in HNC correlate with reduced survival, patients’ response to the EGFR inhibitor cetuximab often declines rapidly after a short period of effectiveness. Hyung Kwon Byeon at Korea University College of Medicine in Seoul and colleagues review current knowledge of the mechanisms underlying cetuximab resistance. They suggest that evaluating a patient’s genetic profile and combining cetuximab with drugs that enhance the effects of inhibiting EGFR signaling pathways (with inhibitors of other EGFR family members or proteins that mediate EGFR entry to the cell nucleus, for example) as well as with agents that inhibit cancer cell metabolism could be a more effective approach for treating HNC.
Collapse
Affiliation(s)
- Hyung Kwon Byeon
- Department of Otorhinolaryngology-Head and Neck Surgery, Soonchunhyang University College of Medicine, Seoul, Republic of Korea. .,Systems Molecular Oncology for Head and Neck Cancer, Seoul, Republic of Korea. .,Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea.
| | - Minhee Ku
- Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea.,Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Research Institute of Radiological Science, Yonsei University, Seoul, Republic of Korea
| | - Jaemoon Yang
- Systems Molecular Radiology at Yonsei, Seoul, Republic of Korea. .,Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Research Institute of Radiological Science, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
252
|
Fu J, Xiong Z, Huang C, Li J, Yang W, Han Y, Paiboonrungruan C, Major MB, Chen KN, Kang X, Chen X. Hyperactivity of the transcription factor Nrf2 causes metabolic reprogramming in mouse esophagus. J Biol Chem 2019; 294:327-340. [PMID: 30409900 PMCID: PMC6322899 DOI: 10.1074/jbc.ra118.005963] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/03/2018] [Indexed: 12/17/2022] Open
Abstract
Mutations in the genes encoding nuclear factor (erythroid-derived 2)-like 2 (NRF2), Kelch-like ECH-associated protein 1 (KEAP1), and cullin 3 (CUL3) are commonly observed in human esophageal squamous cell carcinoma (ESCC) and result in activation of the NRF2 signaling pathway. Moreover, hyperactivity of the transcription factor Nrf2 has been found to cause esophageal hyperproliferation and hyperkeratosis in mice. However, the underlying mechanism is unclear. In this study, we aimed to understand the molecular mechanisms of esophageal hyperproliferation in mice due to hyperactive Nrf2. Esophageal tissues were obtained from genetically modified mice that differed in the status of the Nrf2 gene and genes in the same pathway (Nrf2-/-, Keap1-/-, K5Cre;Pkm2fl/fl;Keap1-/-, and WT) and analyzed for metabolomic profiles, Nrf2 ChIP-seq, and gene expression. We found that hyperactive Nrf2 causes metabolic reprogramming and up-regulation of metabolic genes in the mouse esophagus. One of the glycolysis genes encoding pyruvate kinase M2 (Pkm2) was not only differentially up-regulated, but also glycosylated and oligomerized, resulting in increased ATP biosynthesis. However, constitutive knockout of Pkm2 failed to inhibit this esophageal phenotype in vivo, and this failure may have been due to compensation by Pkm1 up-regulation. Transient inhibition of NRF2 or glycolysis inhibited the growth of human ESCC cells in which NRF2 is hyperactive in vitro In summary, hyperactive Nrf2 causes metabolic reprogramming in the mouse esophagus through its transcriptional regulation of metabolic genes. Blocking glycolysis transiently inhibits cell proliferation and may therefore have therapeutically beneficial effects on NRF2high ESCC in humans.
Collapse
Affiliation(s)
- Junsheng Fu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province 350002, China; Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Zhaohui Xiong
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Caizhi Huang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Jing Li
- Department of Thoracic Surgery, Ningxia Medical University General Hospital, Yinchuan, Ningxia 750004, China
| | - Wenjun Yang
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), Cancer Institute of the General Hospital, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Yuning Han
- Department of Thoracic Surgery, Ningxia Medical University General Hospital, Yinchuan, Ningxia 750004, China
| | - Chorlada Paiboonrungruan
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707
| | - Michael B Major
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Ke-Neng Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaozheng Kang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Xiaoxin Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina 27707; Center for Esophageal Disease and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599.
| |
Collapse
|
253
|
By reducing global mRNA translation in several ways, 2-deoxyglucose lowers MCL-1 protein and sensitizes hemopoietic tumor cells to BH3 mimetic ABT737. Cell Death Differ 2018; 26:1766-1781. [PMID: 30538285 PMCID: PMC6748140 DOI: 10.1038/s41418-018-0244-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/30/2018] [Accepted: 11/15/2018] [Indexed: 01/15/2023] Open
Abstract
Drugs targeting various pro-survival BCL-2 family members (‘‘BH3 mimetics’’) have efficacy in hemopoietic malignancies, but the non-targeted pro-survival family members can promote resistance. Pertinently, the sensitivity of some tumor cell lines to BH3 mimetic ABT737, which targets BCL-2, BCL-XL, and BCL-W but not MCL-1, is enhanced by 2-deoxyglucose (2DG). We found that 2DG augmented apoptosis induced by ABT737 in 3 of 8 human hemopoietic tumor cell lines, most strongly in pre-B acute lymphocytic leukemia cell line NALM-6, the focus of our mechanistic studies. Although 2DG can lower MCL-1 translation, how it does so is incompletely understood, in part because 2DG inhibits both glycolysis and protein glycosylation in the endoplasmic reticulum (ER). Its glycolysis inhibition lowered ATP and, through the AMPK/mTORC1 pathway, markedly reduced global protein synthesis, as did an ER integrated stress response. A dual reporter assay revealed that 2DG impeded not only cap-dependent translation but also elongation or cap-independent translation. MCL-1 protein fell markedly, whereas 12 other BCL-2 family members were unaffected. We ascribe the MCL-1 drop to the global fall in translation, exacerbated for mRNAs with a structured 5′ untranslated region (5′UTR) containing potential regulatory motifs like those in MCL-1 mRNA and the short half-life of MCL-1 protein. Pertinently, 2DG downregulated two other short-lived oncoproteins, MYC and MDM2. Thus, our results support MCL-1 as a critical 2DG target, but also reveal multiple effects on global translation that may well also affect its promotion of apoptosis.
Collapse
|
254
|
Guerra AR, Duarte MF, Duarte IF. Targeting Tumor Metabolism with Plant-Derived Natural Products: Emerging Trends in Cancer Therapy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10663-10685. [PMID: 30227704 DOI: 10.1021/acs.jafc.8b04104] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Recognition of neoplastic metabolic reprogramming as one of cancer's hallmarks has paved the way for developing novel metabolism-targeted therapeutic approaches. The use of plant-derived natural bioactive compounds for this endeavor is especially promising, due to their diverse structures and multiple targets. Hence, over the past decade, a growing number of studies have assessed the impact of phytochemicals on tumor cell metabolism, aiming at improving current knowledge on their mechanisms of action and, at the same time, evaluating their potential as anti-cancer metabolic modulators. In this Review, we focus on three classes of plant-derived compounds with promising anti-cancer activity-phenolic compounds, isoprenoids, and alkaloids-to describe their effects on major energetic and biosynthetic pathways of human tumor cells. Such a comprehensive and integrated account of the ability of these compounds to hit different metabolic targets is expected to contribute to the rational design and critical assessment of novel anti-cancer therapies based on natural-product-mediated metabolic reprogramming.
Collapse
Affiliation(s)
- Angela R Guerra
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja , Apartado 6158 , 7801-908 Beja , Portugal
- CICECO - Instituto de Materiais de Aveiro, Departamento de Quı́mica , Universidade de Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| | - Maria F Duarte
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja , Apartado 6158 , 7801-908 Beja , Portugal
- ICAAM - Instituto de Ciências Agrárias e Ambientais Mediterrânicas , Universidade de Évora , Pólo da Mitra, 7006-554 Évora , Portugal
| | - Iola F Duarte
- CICECO - Instituto de Materiais de Aveiro, Departamento de Quı́mica , Universidade de Aveiro , Campus de Santiago , 3810-193 Aveiro , Portugal
| |
Collapse
|
255
|
Koenig JB, Dulla CG. Dysregulated Glucose Metabolism as a Therapeutic Target to Reduce Post-traumatic Epilepsy. Front Cell Neurosci 2018; 12:350. [PMID: 30459556 PMCID: PMC6232824 DOI: 10.3389/fncel.2018.00350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of disability worldwide and can lead to post-traumatic epilepsy. Multiple molecular, cellular, and network pathologies occur following injury which may contribute to epileptogenesis. Efforts to identify mechanisms of disease progression and biomarkers which predict clinical outcomes have focused heavily on metabolic changes. Advances in imaging approaches, combined with well-established biochemical methodologies, have revealed a complex landscape of metabolic changes that occur acutely after TBI and then evolve in the days to weeks after. Based on this rich clinical and preclinical data, combined with the success of metabolic therapies like the ketogenic diet in treating epilepsy, interest has grown in determining whether manipulating metabolic activity following TBI may have therapeutic value to prevent post-traumatic epileptogenesis. Here, we focus on changes in glucose utilization and glycolytic activity in the brain following TBI and during seizures. We review relevant literature and outline potential paths forward to utilize glycolytic inhibitors as a disease-modifying therapy for post-traumatic epilepsy.
Collapse
Affiliation(s)
- Jenny B Koenig
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
256
|
Angelopoulou A, Kolokithas-Ntoukas A, Papaioannou L, Kakazanis Z, Khoury N, Zoumpourlis V, Papatheodorou S, Kardamakis D, Bakandritsos A, Hatziantoniou S, Avgoustakis K. Canagliflozin-loaded magnetic nanoparticles as potential treatment of hypoxic tumors in combination with radiotherapy. Nanomedicine (Lond) 2018; 13:2435-2454. [PMID: 30311542 DOI: 10.2217/nnm-2018-0145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM To synthesize magnetic nanoparticles loaded with the SGLT2-inhibitor canagliflozin (CANA) and evaluate its anticancer potential under normoxic and hypoxic conditions in combination or not with radiotherapy. MATERIAL & METHODS Iron oxide nanoparticles were synthesized via an alkaline hydrolytic precipitation of iron precursor in the presence of poly(methacrylic acid)-graft-poly(ethyleneglycol methacrylate). CANA was conjugated to the nanoparticles using N-ethyl-N'-(3-dimethyl aminopropyl) carbodiimide (EDC)/N-hydroxysuccinimide chemistry. The anticancer efficacy of the nanoparticles was evaluated in cancer cell lines and in a mouse PDV C57 tumor model. RESULTS In the mouse xenograft cancer model, the combination of CANA-loaded nanoparticles with radiotherapy (in the presence of an external magnetic field at the tumor site) exhibited higher antitumor activity compared with the combination of free CANA with radiotherapy. CONCLUSION The results obtained indicate the potential that the combination of selective delivery of a SGLT2 inhibitor such as CANA with radiotherapy holds as an anticancer treatment.
Collapse
Affiliation(s)
- Athina Angelopoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece
| | | | - Ligeri Papaioannou
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece
| | - Zacharias Kakazanis
- Institute of Biology, Medicinal Chemistry & Biotechnology, NHRF, Athens, Greece
| | - Nikolas Khoury
- Institute of Biology, Medicinal Chemistry & Biotechnology, NHRF, Athens, Greece
| | | | | | - Dimitrios Kardamakis
- Department of Radiation Oncology, School of Health Sciences, University of Patras, Patras 26504, Greece
| | - Aristides Bakandritsos
- Department of Physical Chemistry, Faculty of Science, Regional Centre for Advanced Technologies & Materials, Palacky University in Olomouc, 17 listopadu 1192/12, 77146 Olomouc, Czech Republic
| | - Sophia Hatziantoniou
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece
| | - Konstantinos Avgoustakis
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece.,Clinical Stidues Unit, Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, Athens 11527, Greece
| |
Collapse
|
257
|
Fortunato S, Bononi G, Granchi C, Minutolo F. An Update on Patents Covering Agents That Interfere with the Cancer Glycolytic Cascade. ChemMedChem 2018; 13:2251-2265. [DOI: 10.1002/cmdc.201800447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/07/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Serena Fortunato
- Dipartimento di FarmaciaUniversità di Pisa Via Bonanno 33 56126 Pisa Italy
| | - Giulia Bononi
- Dipartimento di FarmaciaUniversità di Pisa Via Bonanno 33 56126 Pisa Italy
| | - Carlotta Granchi
- Dipartimento di FarmaciaUniversità di Pisa Via Bonanno 33 56126 Pisa Italy
| | - Filippo Minutolo
- Dipartimento di FarmaciaUniversità di Pisa Via Bonanno 33 56126 Pisa Italy
| |
Collapse
|
258
|
Lucantoni F, Dussmann H, Prehn JHM. Metabolic Targeting of Breast Cancer Cells With the 2-Deoxy-D-Glucose and the Mitochondrial Bioenergetics Inhibitor MDIVI-1. Front Cell Dev Biol 2018; 6:113. [PMID: 30255019 PMCID: PMC6141706 DOI: 10.3389/fcell.2018.00113] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023] Open
Abstract
Breast cancer cells have different requirements on metabolic pathways in order to sustain their growth. Triple negative breast cancer (TNBC), an aggressive breast cancer subtype relies mainly on glycolysis, while estrogen receptor positive (ER+) breast cancer cells possess higher mitochondrial oxidative phosphorylation (OXPHOS) levels. However, breast cancer cells generally employ both pathways to sustain their metabolic needs and to compete with the surrounding environment. In this study, we demonstrate that the mitochondrial fission inhibitor MDIVI-1 alters mitochondrial bioenergetics, at concentrations that do not affect mitochondrial morphology. We show that this effect is accompanied by an increase in glycolysis consumption. Dual targeting of glycolysis with 2-deoxy-D-glucose (2DG) and mitochondrial bioenergetics with MDIVI-1 reduced cellular bioenergetics, increased cell death and decreased clonogenic activity of MCF7 and HDQ-P1 breast cancer cells. In conclusion, we have explored a novel and effective combinatorial regimen for the treatment of breast cancer.
Collapse
Affiliation(s)
- Federico Lucantoni
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
259
|
Wilson JJ, Chow KH, Labrie NJ, Branca JA, Sproule TJ, Perkins BRA, Wolf EE, Costa M, Stafford G, Rosales C, Mills KD, Roopenian DC, Hasham MG. Enhancing the efficacy of glycolytic blockade in cancer cells via RAD51 inhibition. Cancer Biol Ther 2018; 20:169-182. [PMID: 30183475 PMCID: PMC6343731 DOI: 10.1080/15384047.2018.1507666] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models – spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells – can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4ʹ-Diisothiocyano-2,2ʹ-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.
Collapse
Affiliation(s)
- John J Wilson
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Kin-Hoe Chow
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Nathan J Labrie
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Jane A Branca
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Thomas J Sproule
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Bryant R A Perkins
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Elise E Wolf
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Mauro Costa
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Grace Stafford
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Christine Rosales
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | | | - Derry C Roopenian
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| | - Muneer G Hasham
- a Research Department , The Jackson Laboratory , Bar Harbor , Maine , USA
| |
Collapse
|
260
|
Shao LR, Rho JM, Stafstrom CE. Glycolytic inhibition: A novel approach toward controlling neuronal excitability and seizures. Epilepsia Open 2018; 3:191-197. [PMID: 30564778 PMCID: PMC6293058 DOI: 10.1002/epi4.12251] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Conventional antiseizure medications reduce neuronal excitability through effects on ion channels or synaptic function. In recent years, it has become clear that metabolic factors also play a crucial role in the modulation of neuronal excitability. Indeed, metabolic regulation of neuronal excitability is pivotal in seizure pathogenesis and control. The clinical effectiveness of a variety of metabolism‐based diets, especially for children with medication‐refractory epilepsy, underscores the applicability of metabolic approaches to the control of seizures and epilepsy. Such diets include the ketogenic diet, the modified Atkins diet, and the low‐glycemic index treatment (among others). A promising avenue to alter cellular metabolism, and hence excitability, is by partial inhibition of glycolysis, which has been shown to reduce seizure susceptibility in a variety of animal models as well as in cellular systems in vitro. One such glycolytic inhibitor, 2‐deoxy‐d‐glucose (2DG), increases seizure threshold in vivo and reduces interictal and ictal epileptiform discharges in hippocampal slices. Here, we review the role of glucose metabolism and glycolysis on neuronal excitability, with specific reference to 2DG, and discuss the potential use of 2DG and similar agents in the clinical arena for seizure management.
Collapse
Affiliation(s)
- Li-Rong Shao
- Division of Pediatric Neurology Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland U.S.A
| | - Jong M Rho
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology Alberta Children's Hospital Research Institute Hotchkiss Brain Institute Cumming School of Medicine University of Calgary Calgary Alberta Canada
| | - Carl E Stafstrom
- Division of Pediatric Neurology Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland U.S.A
| |
Collapse
|
261
|
Turetta M, Bulfoni M, Brisotto G, Fasola G, Zanello A, Biscontin E, Mariuzzi L, Steffan A, Di Loreto C, Cesselli D, Del Ben F. Assessment of the Mutational Status of NSCLC Using Hypermetabolic Circulating Tumor Cells. Cancers (Basel) 2018; 10:cancers10080270. [PMID: 30110953 PMCID: PMC6115779 DOI: 10.3390/cancers10080270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/01/2018] [Accepted: 08/10/2018] [Indexed: 01/11/2023] Open
Abstract
Molecular characterization is currently a key step in NSCLC therapy selection. Circulating tumor cells (CTC) are excellent candidates for downstream analysis, but technology is still lagging behind. In this work, we show that the mutational status of NSCLC can be assessed on hypermetabolic CTC, detected by their increased glucose uptake. We validated the method in 30 Stage IV NSCLC patients: peripheral blood samples were incubated with a fluorescent glucose analog (2-NBDG) and analyzed by flow cytometry. Cells with the highest glucose uptake were sorted out. EGFR and KRAS mutations were detected by ddPCR. In sorted cells, mutated DNA was found in 85% of patients, finding an exact match with primary tumor in 70% of cases. Interestingly, in two patients multiple KRAS mutations were detected. Two patients displayed different mutations with respect to the primary tumor, and in two out of the four patients with a wild type primary tumor, new mutations were highlighted: EGFR p.746_750del and KRAS p.G12V. Hypermetabolic CTC can be enriched without the need of dedicated equipment and their mutational status can successfully be assessed by ddPCR. Finally, the finding of new mutations supports the possibility of probing tumor heterogeneity.
Collapse
Affiliation(s)
- Matteo Turetta
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
| | - Michela Bulfoni
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers, C.R.O. Aviano National Cancer Institute IRCCS, via F. Gallini 2, 33081 Aviano (PN), Italy.
- IOV-IRCCS, Immunology and Molecular Oncology Unit, V. Gattamelata 64, 35128 Padova, Italy.
- DISCOG, University of Padova, V. Giustiniani 2, 35128 Padova, Italy.
| | - Gianpiero Fasola
- Udine Academic Hospital, P.le Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Andrea Zanello
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
| | - Eva Biscontin
- Immunopathology and Cancer Biomarkers, C.R.O. Aviano National Cancer Institute IRCCS, via F. Gallini 2, 33081 Aviano (PN), Italy.
| | - Laura Mariuzzi
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
- Udine Academic Hospital, P.le Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, C.R.O. Aviano National Cancer Institute IRCCS, via F. Gallini 2, 33081 Aviano (PN), Italy.
| | - Carla Di Loreto
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
- Udine Academic Hospital, P.le Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Daniela Cesselli
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
- Udine Academic Hospital, P.le Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Fabio Del Ben
- Department of Medicine, University of Udine, P.le Kolbe 4, 33100 Udine, Italy.
- Immunopathology and Cancer Biomarkers, C.R.O. Aviano National Cancer Institute IRCCS, via F. Gallini 2, 33081 Aviano (PN), Italy.
| |
Collapse
|
262
|
Carreño D, Corro N, Torres-Estay V, Véliz LP, Jaimovich R, Cisternas P, San Francisco IF, Sotomayor PC, Tanasova M, Inestrosa NC, Godoy AS. Fructose and prostate cancer: toward an integrated view of cancer cell metabolism. Prostate Cancer Prostatic Dis 2018; 22:49-58. [PMID: 30104655 DOI: 10.1038/s41391-018-0072-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/13/2018] [Accepted: 06/29/2018] [Indexed: 01/07/2023]
Abstract
Activation of glucose transporter-1 (Glut-1) gene expression is a molecular feature of cancer cells that increases glucose uptake and metabolism. Increased glucose uptake is the basis for the clinical localization of primary tumors using positron emission tomography (PET) and 2-deoxy-2-[18F]-fluoro-D-glucose (FDG) as a radiotracer. However, previous studies have demonstrated that a considerable number of cancers, which include prostate cancer (CaP), express low to undetectable levels of Glut-1 and that FDG-PET has limited clinical applicability in CaP. This observation could be explained by a low metabolic activity of CaP cells that may be overcome using different hexoses, such as fructose, as the preferred energy source. However, these hypotheses have not been examined critically in CaP. This review article summarizes what is currently known about transport and metabolism of hexoses, and more specifically fructose, in CaP and provides experimental evidences indicating that CaP cells may have increased capacity to transport and metabolize fructose in vitro and in vivo. Moreover, this review highlights recent findings that allow better understanding of how metabolism of fructose may regulate cancer cell proliferation and how fructose uptake and metabolism, through the de novo lipogenesis pathway, may provide new opportunities for CaP early diagnosis, staging, and treatment.
Collapse
Affiliation(s)
- Daniela Carreño
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Néstor Corro
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Loreto P Véliz
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE), Department of Cell Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Paula C Sotomayor
- Center for Integrative Medicine and Innovative Science, Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Marina Tanasova
- Department of Chemistry, Michigan Technological University, Houghton, MI, 49931, USA
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Department of Cell Biology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro S Godoy
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
263
|
Pesi R, Petrotto E, Colombaioni L, Allegrini S, Garcia-Gil M, Camici M, Jordheim LP, Tozzi MG. Cytosolic 5'-Nucleotidase II Silencing in a Human Lung Carcinoma Cell Line Opposes Cancer Phenotype with a Concomitant Increase in p53 Phosphorylation. Int J Mol Sci 2018; 19:E2115. [PMID: 30037008 PMCID: PMC6073589 DOI: 10.3390/ijms19072115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022] Open
Abstract
Purine homeostasis is maintained by a purine cycle in which the regulated member is a cytosolic 5'-nucleotidase II (cN-II) hydrolyzing IMP and GMP. Its expression is particularly high in proliferating cells, indeed high cN-II activity or expression in hematological malignancy has been associated to poor prognosis and chemoresistance. Therefore, a strong interest has grown in developing cN-II inhibitors, as potential drugs alone or in combination with other compounds. As a model to study the effect of cN-II inhibition we utilized a lung carcinoma cell line (A549) in which the enzyme was partially silenced and its low activity conformation was stabilized through incubation with 2-deoxyglucose. We measured nucleotide content, reduced glutathione, activities of enzymes involved in glycolysis and Krebs cycle, protein synthesis, mitochondrial function, cellular proliferation, migration and viability. Our results demonstrate that high cN-II expression is associated with a glycolytic, highly proliferating phenotype, while silencing causes a reduction of proliferation, protein synthesis and migration ability, and an increase of oxidative performances. Similar results were obtained in a human astrocytoma cell line. Moreover, we demonstrate that cN-II silencing is concomitant with p53 phosphorylation, suggesting a possible involvement of this pathway in mediating some of cN-II roles in cancer cell biology.
Collapse
Affiliation(s)
- Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy.
| | - Edoardo Petrotto
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy.
| | - Laura Colombaioni
- Istituto di Neuroscienze, CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy.
| | - Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy.
| | - Mercedes Garcia-Gil
- Unità Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy.
| | - Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy.
| | - Lars Petter Jordheim
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France.
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy.
| |
Collapse
|
264
|
Ishino K, Kudo M, Peng WX, Kure S, Kawahara K, Teduka K, Kawamoto Y, Kitamura T, Fujii T, Yamamoto T, Wada R, Naito Z. 2-Deoxy- d -glucose increases GFAT1 phosphorylation resulting in endoplasmic reticulum-related apoptosis via disruption of protein N -glycosylation in pancreatic cancer cells. Biochem Biophys Res Commun 2018; 501:668-673. [DOI: 10.1016/j.bbrc.2018.05.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/07/2018] [Indexed: 12/28/2022]
|
265
|
Overexpression of miR-202 resensitizes imatinib resistant chronic myeloid leukemia cells through targetting Hexokinase 2. Biosci Rep 2018; 38:BSR20171383. [PMID: 29559564 PMCID: PMC5938424 DOI: 10.1042/bsr20171383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 03/06/2018] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease which uniquely expresses a constitutively active tyrosine kinase, BCR/ABL. As a specific inhibitor of the BCR-ABL tyrosine kinase, imatinib becomes the first choice for the treatment of CML due to its high efficacy and low toxicity. However, the development of imatinib resistance limits the long-term treatment benefits of it in CML patients. In the present study, we aimed to investigate the roles of miR-202 in the regulation of imatinib sensitivity in CML cell lines and the possible mechanisms involved in this process. We found miR-202 was down-regulated in seven CML cell lines by quantitative reverse-transcription PCR (qRT-PCR) analysis. Overexpression of miR-202 significantly suppressed proliferation rates of CML cells. By establishing imatinib resistant cell lines originating from K562 and KU812 cells, we observed expressions of miR-202 were down-regulated by imatinib treatments and imatinib resistant CML cell lines exhibited lower level of miR-202. On the contrary, imatinib resistant CML cell lines displayed up-regulated glycolysis rate than sensitive cells with the evidence that glucose uptake, lactate production, and key glycolysis enzymes were elevated in imatinib resistant cells. Importantly, the imatinib resistant CML cell lines were more sensitive to glucose starvation and glycolysis inhibitors. In addition, we identified Hexokinase 2 (HK2) as a direct target of miR-202 in CML cell lines. Overexpression of miR-202 sensitized imatinib resistant CML through the miR-202-mediated glycolysis inhibition by targetting HK2. Finally, we provided the clinical relevance that miR-202 was down-regulated in CML patients and patients with lower miR-202 expression displayed higher HK2 expression. The present study will provide new aspects on the miRNA-modulated tyrosine kinase inhibitor (TKI) sensitivity in CML, contributing to the development of new therapeutic anticancer drugs.
Collapse
|
266
|
Propranolol sensitizes prostate cancer cells to glucose metabolism inhibition and prevents cancer progression. Sci Rep 2018; 8:7050. [PMID: 29728578 PMCID: PMC5935740 DOI: 10.1038/s41598-018-25340-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/18/2018] [Indexed: 02/04/2023] Open
Abstract
Propranolol, a widely used non-selective beta-adrenergic receptor blocker, was recently shown to display anticancer properties. Its potential to synergize with certain drugs has been also outlined. However, it is necessary to take into account all the properties of propranolol to select a drug that could be efficiently combined with. Propranolol was reported to block the late phase of autophagy. Hence, we hypothesized that in condition enhancing autophagy flux, cancer cells should be especially sensitive to propranolol. 2DG, a glycolysis inhibitor, is an anti-tumor agent having limited effect in monotherapy notably due to induction of pro-survival autophagy. Here, we report that treatment of cancer cells with propranolol in combination with the glycolysis inhibitor 2DG induced a massive accumulation of autophagosome due to autophagy blockade. The propranolol +2DG treatment efficiently prevents prostate cancer cell proliferation, induces cell apoptosis, alters mitochondrial morphology, inhibits mitochondrial bioenergetics and aggravates ER stress in vitro and also suppresses tumor growth in vivo. Our study underlines for the first time the interest to take advantage of the ability of propranolol to inhibit autophagy to design new anti-cancer therapies.
Collapse
|
267
|
Keating E, Martel F. Antimetabolic Effects of Polyphenols in Breast Cancer Cells: Focus on Glucose Uptake and Metabolism. Front Nutr 2018; 5:25. [PMID: 29713632 PMCID: PMC5911477 DOI: 10.3389/fnut.2018.00025] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/27/2018] [Indexed: 12/20/2022] Open
Abstract
In the last years, metabolic reprogramming became a new key hallmark of tumor cells. One of its components is a deviant energetic metabolism, known as Warburg effect—an aerobic lactatogenesis—characterized by elevated rates of glucose uptake and consumption with high-lactate production even in the presence of oxygen. Because many cancer cells display a greater sensitivity to glucose deprivation-induced cytotoxicity than normal cells, inhibitors of glucose cellular uptake (facilitative glucose transporter 1 inhibitors) and oxidative metabolism (glycolysis inhibitors) are potential therapeutic targets in cancer treatment. Polyphenols, abundantly contained in fruits and vegetables, are dietary components with an established protective role against cancer. Several molecular mechanisms are involved in the anticancer effect of polyphenols, including effects on apoptosis, cell cycle regulation, plasma membrane receptors, signaling pathways, and epigenetic mechanisms. Additionally, inhibition of glucose cellular uptake and metabolism in cancer cell lines has been described for several polyphenols, and this effect was shown to be associated with their anticarcinogenic effect. This work will review data showing an antimetabolic effect of polyphenols and its involvement in the chemopreventive/chemotherapeutic potential of these dietary compounds, in relation to breast cancer.
Collapse
Affiliation(s)
- Elisa Keating
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal.,CINTESIS, Center for Research in Health Technologies and Information Systems, University of Porto, Porto, Portugal
| | - Fátima Martel
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
268
|
El Arfani C, De Veirman K, Maes K, De Bruyne E, Menu E. Metabolic Features of Multiple Myeloma. Int J Mol Sci 2018; 19:E1200. [PMID: 29662010 PMCID: PMC5979361 DOI: 10.3390/ijms19041200] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/07/2018] [Accepted: 04/10/2018] [Indexed: 01/19/2023] Open
Abstract
Cancer is known for its cellular changes contributing to tumour growth and cell proliferation. As part of these changes, metabolic rearrangements are identified in several cancers, including multiple myeloma (MM), which is a condition whereby malignant plasma cells accumulate in the bone marrow (BM). These metabolic changes consist of generation, inhibition and accumulation of metabolites and metabolic shifts in MM cells. Changes in the BM micro-environment could be the reason for such adjustments. Enhancement of glycolysis and glutaminolysis is found in MM cells compared to healthy cells. Metabolites and enzymes can be upregulated or downregulated and play a crucial role in drug resistance. Therefore, this review will focus on changes in glucose and glutamine metabolism linked with the emergence of drug resistance. Moreover, metabolites do not only affect other metabolic components to benefit cancer development; they also interfere with transcription factors involved in proliferation and apoptotic regulation.
Collapse
Affiliation(s)
- Chaima El Arfani
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| |
Collapse
|
269
|
Gao L, Wang KX, Zhang NN, Li JQ, Qin XM, Wang XL. 1H Nuclear Magnetic Resonance Based Metabolomics Approach Reveals the Metabolic Mechanism of (−)-5-Hydroxy-equol against Hepatocellular Carcinoma Cells in Vitro. J Proteome Res 2018; 17:1833-1843. [DOI: 10.1021/acs.jproteome.7b00853] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
| | - Ke-xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
- College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Nan-nan Zhang
- College of Life Sciences, Hebei Agricultural University, Baoding 071001, PR China
| | - Jia-qi Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
- College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Xue-mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
| | - Xiu-ling Wang
- College of Life Sciences, Hebei Agricultural University, Baoding 071001, PR China
| |
Collapse
|
270
|
Chen J, Hu X, Cui J. Shikonin, vitamin K 3 and vitamin K 5 inhibit multiple glycolytic enzymes in MCF-7 cells. Oncol Lett 2018; 15:7423-7432. [PMID: 29725454 DOI: 10.3892/ol.2018.8251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Glycolysis is the most important source of energy for the production of anabolic building blocks in cancer cells. Therefore, glycolytic enzymes are regarded as potential targets for cancer treatment. Previously, naphthaquinones, including shikonin, vitamin K3 and vitamin K5, have been proven to decrease the rate of glycolysis in cancer cells, which is partly due to suppressed pyruvate kinase activity. In the present study, enzymatic assays were performed using MCF-7 cell lysate in order to screen the profile of glycolytic enzymes in cancer cells inhibited by shikonin, vitamin K3 and vitamin K5, in addition to pyruvate kinase. Results revealed that hexokinase, phosphofructokinase-1, fructose bisphosphate aldolase, glyceraldehyde-3-phosphate dehydrogenase and pyruvate kinase produced in the process of glycolysis were inhibited by shikonin, vitamin K3 and vitamin K5. The results indicated that shikonin, vitamin K3 and vitamin K5 are chemical inhibitors of glycolytic enzymes in cancer cells and have potential uses in translational medical applications.
Collapse
Affiliation(s)
- Jing Chen
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 310036, P.R. China
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Prevention and Intervention, National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310009, P.R. China
| | - Jingjie Cui
- Department of Biomedical Engineering, College of Life Information Science and Instrument Engineering, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
271
|
Bonuccelli G, Peiris-Pages M, Ozsvari B, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Targeting cancer stem cell propagation with palbociclib, a CDK4/6 inhibitor: Telomerase drives tumor cell heterogeneity. Oncotarget 2018; 8:9868-9884. [PMID: 28039467 PMCID: PMC5354777 DOI: 10.18632/oncotarget.14196] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/16/2016] [Indexed: 12/24/2022] Open
Abstract
In this report, we systematically examined the role of telomerase activity in lung and ovarian cancer stem cell (CSC) propagation. For this purpose, we indirectly gauged telomerase activity, by linking the hTERT-promoter to eGFP. Using lung (A549) and ovarian (SKOV3) cancer cells, transduced with the hTERT-GFP reporter, we then employed GFP-expression levels to fractionate these cell lines into GFP-high and GFP-low populations. We functionally compared the phenotype of these GFP-high and GFP-low populations. More specifically, we now show that the cancer cells with higher telomerase activity (GFP-high) are more energetically activated, with increased mitochondrial mass and function, as well as increased glycolytic activity. This was further validated and confirmed by unbiased proteomics analysis. Cells with high telomerase activity also showed an increased capacity for stem cell activity (as measured using the 3D-spheroid assay) and cell migration (as measured using a Boyden chamber approach). These enhanced biological phenotypes were effectively inhibited by classical modulators of energy metabolism, which target either i) mitochondrial metabolism (i.e., oligomycin) or ii) glycolysis (i.e., 2-deoxy-glucose), or iii) by using the FDA-approved antibiotic doxycycline, which inhibits mitochondrial biogenesis. Finally, the level of telomerase activity also determined the ability of hTERT-high cells to proliferate, as assessed by measuring DNA synthesis via EdU incorporation. Consistent with these observations, treatment with an FDA-approved CDK4/6 inhibitor (PD-0332991/palbociclib) specifically blocked the propagation of both lung and ovarian CSCs. Virtually identical results were obtained with breast CSCs, which were also highly sensitive to palbociclib at concentrations in the nanomolar range. In summary, CSCs with high telomerase activity are among the most energetically activated, migratory and proliferative cell sub-populations. These observations may provide a mechanistic explanation for tumor metabolic heterogeneity, based on telomerase activity. FDA-approved drugs, such as doxycycline and palbociclib, were both effective at curtailing CSC propagation. Thus, these FDA-approved drugs could be used to target telomerase-high proliferative CSCs, in multiple cancer types. Finally, our experiments also allowed us to distinguish two different cellular populations of hTERT-high cells, one that was proliferative (i.e., replicative immortality) and the other that was non-proliferative (i.e., quiescent). We speculate that the non-proliferative population of hTERT-high cells that we identified could be mechanistically involved in tumor dormancy.
Collapse
Affiliation(s)
- Gloria Bonuccelli
- Paterson Building, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Maria Peiris-Pages
- Paterson Building, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Bela Ozsvari
- Paterson Building, University of Manchester, Manchester M20 4BX, United Kingdom
| | | | - Federica Sotgia
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC), University of Salford, Greater Manchester, M5 4WT, United Kingdom
| | - Michael P Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre (BRC), University of Salford, Greater Manchester, M5 4WT, United Kingdom
| |
Collapse
|
272
|
Halicka HD, Garcia J, Li J, Zhao H, Darzynkiewicz Z. Synergy of 2-deoxy-D-glucose combined with berberine in inducing the lysosome/autophagy and transglutaminase activation-facilitated apoptosis. Apoptosis 2018; 22:229-238. [PMID: 27796611 DOI: 10.1007/s10495-016-1315-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Utilizing a variety of flow cytometric methods evidence was obtained indicating that a combination of the glucose analog 2-deoxy-D-glucose (2-dG) and the plant alkaloid berberine (BRB) produces synergistic effect in the induction of apoptosis in human lymphoblastoid TK6 cells. The synergistic effect is seen at concentrations of the drugs at which each of them alone shows no cytotoxicity at all. The data suggest that the combination of these drugs, which are known in terms of their overall toxicity, side effects and pharmacokinetics may be considered for further studies as chemopreventive and cancer treatment modalities. Of interest are results indicating that rapamycin, which similarly to BRB, suppresses mTOR signaling, when combined with 2-dG shows no synergistic properties. Metformin, on other hand, requires much higher concentration to show the synergy with 2-dG. Also of interest are the findings pertaining to the methodology of the present study. Specifically, dynamic assessment of cellular viability was performed by using the DRAQ7 cell exclusion fluorochrome present in cultures from 0 to 72 h. Concurrent measurement of lysosomal proton pump using acridine orange as the probe shows activation of lysosomes in the cells treated with 2-dG or BRB alone as well as with the drugs combined. Apoptosis was assessed by measuring DNA fragmentation, cell cycle, activation of caspase-3 and tissue transglutaminase (Tgase). A novel cytometric method was developed based on analysis of lysosomal (acidic vesicles) proton pump in live cells followed by cell lysis with detergent and fluorochrome labeling of proteins and DNA to analyze Tgase activation concurrently with cell cycle, in same population of cells. The data show that the cell subpopulation undergoing apoptosis has increased side (right-angle) light scatter likely due to the presence of the crosslinked (solid state) proteins, the consequence Tgase activation.
Collapse
Affiliation(s)
- H Dorota Halicka
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, BSB 438, Valhalla, NY, 10595, USA
| | - Jorge Garcia
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, BSB 438, Valhalla, NY, 10595, USA
| | - Jiangwei Li
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, BSB 438, Valhalla, NY, 10595, USA
| | - Hong Zhao
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, BSB 438, Valhalla, NY, 10595, USA
| | - Zbigniew Darzynkiewicz
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, BSB 438, Valhalla, NY, 10595, USA.
| |
Collapse
|
273
|
Angelopoulou A, Voulgari E, Kolokithas-Ntoukas A, Bakandritsos A, Avgoustakis K. Magnetic Nanoparticles for the Delivery of Dapagliflozin to Hypoxic Tumors: Physicochemical Characterization and Cell Studies. AAPS PharmSciTech 2018; 19:621-633. [PMID: 28924948 DOI: 10.1208/s12249-017-0874-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/05/2017] [Indexed: 11/30/2022] Open
Abstract
In solid tumors, hypoxia (lack of oxygen) is developed, which leads to the development of resistance of tumor cells to chemotherapy and radiotherapy through various mechanisms. Nevertheless, hypoxic cells are particularly vulnerable when glycolysis is inhibited. For this reason, in this study, the development of magnetically targetable nanocarriers of the sodium-glucose transporter protein (SGLT2) inhibitor dapagliflozin (DAPA) was developed for the selective delivery of DAPA in tumors. This nanomedicine in combination with radiotherapy or chemotherapy should be useful for effective treatment of hypoxic tumors. The magnetic nanoparticles consisted of a magnetic iron oxide core and a poly(methacrylic acid)-graft-poly(ethyleneglycol methacrylate) (PMAA-g-PEGMA) polymeric shell. The drug (dapagliflozin) molecules were conjugated on the surface of these nanoparticles via in vivo hydrolysable ester bonds. The nanoparticles had an average size of ~ 70 nm and exhibited a DAPA loading capacity 10.75% (w/w) for a theoretical loading 21.68% (w/w). The magnetic responsiveness of the nanoparticles was confirmed with magnetophoresis experiments. The dapagliflozin-loaded magnetic nanoparticles exhibited excellent colloidal stability in aqueous and biological media. Minimal (less than 15% in 24 h) drug release from the nanoparticles occurred in physiological pH 7.4; however, drug release was significantly accelerated in pH 5.5. Drug release was also accelerated (triggered) under the influence of an alternating magnetic field. The DAPA-loaded nanoparticles exhibited higher in vitro anticancer activity (cytotoxicity) against A549 human lung cancer cells than free DAPA. The application of an external magnetic field gradient increased the uptake of nanoparticles by cells, leading to increased cytotoxicity. The results justify further in vivo studies of the suitability of DAPA-loaded magnetic nanoparticles for the treatment of hypoxic tumors.
Collapse
|
274
|
Netea-Maier RT, Smit JW, Netea MG. Metabolic changes in tumor cells and tumor-associated macrophages: A mutual relationship. Cancer Lett 2018; 413:102-109. [DOI: 10.1016/j.canlet.2017.10.037] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 12/21/2022]
|
275
|
Ždralević M, Marchiq I, de Padua MMC, Parks SK, Pouysségur J. Metabolic Plasiticy in Cancers-Distinct Role of Glycolytic Enzymes GPI, LDHs or Membrane Transporters MCTs. Front Oncol 2017; 7:313. [PMID: 29326883 PMCID: PMC5742324 DOI: 10.3389/fonc.2017.00313] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023] Open
Abstract
Research on cancer metabolism has recently re-surfaced as a major focal point in cancer field with a reprogrammed metabolism no longer being considered as a mere consequence of oncogenic transformation, but as a hallmark of cancer. Reprogramming metabolic pathways and nutrient sensing is an elaborate way by which cancer cells respond to high bioenergetic and anabolic demands during tumorigenesis. Thus, inhibiting specific metabolic pathways at defined steps should provide potent ways of arresting tumor growth. However, both animal models and clinical observations have revealed that this approach is seriously limited by an extraordinary cellular metabolic plasticity. The classical example of cancer metabolic reprogramming is the preference for aerobic glycolysis, or Warburg effect, where cancers increase their glycolytic flux and produce lactate regardless of the presence of the oxygen. This allows cancer cells to meet the metabolic requirements for high rates of proliferation. Here, we discuss the benefits and limitations of disrupting fermentative glycolysis for impeding tumor growth at three levels of the pathway: (i) an upstream block at the level of the glucose-6-phosphate isomerase (GPI), (ii) a downstream block at the level of lactate dehydrogenases (LDH, isoforms A and B), and (iii) the endpoint block preventing lactic acid export (MCT1/4). Using these examples of genetic disruption targeting glycolysis studied in our lab, we will discuss the responses of different cancer cell lines in terms of metabolic rewiring, growth arrest, and tumor escape and compare it with the broader literature.
Collapse
Affiliation(s)
- Maša Ždralević
- Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, University Côte d’Azur, Nice, France
| | - Ibtissam Marchiq
- Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, University Côte d’Azur, Nice, France
| | - Monique M. Cunha de Padua
- Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, University Côte d’Azur, Nice, France
| | - Scott K. Parks
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
| | - Jacques Pouysségur
- Institute for Research on Cancer and Aging (IRCAN), CNRS, INSERM, Centre A. Lacassagne, University Côte d’Azur, Nice, France
- Medical Biology Department, Centre Scientifique de Monaco (CSM), Monaco, Monaco
| |
Collapse
|
276
|
Důra M, Němejcová K, Jakša R, Bártů M, Kodet O, Tichá I, Michálková R, Dundr P. Expression of Glut-1 in Malignant Melanoma and Melanocytic Nevi: an Immunohistochemical Study of 400 Cases. Pathol Oncol Res 2017; 25:361-368. [PMID: 29128957 DOI: 10.1007/s12253-017-0363-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/08/2017] [Indexed: 11/26/2022]
Abstract
The glucose transporter-1 (Glut-1) is a cell membrane glycoprotein involved in glucose uptake. An increased expression of Glut-1 is an important cell adaptation mechanism against hypoxia. An upregulation of Glut-1 can be found in several types of malignant tumors, which are able to reprogram their metabolism from oxidative phosphorylation to aerobic glycolysis (Warburg effect). However, the data regarding melanocytic lesions is equivocal. We performed comprehensive immunohistochemical analysis of the Glut-1 expression in 225 malignant melanomas (MM) and 175 benign nevi. Only the membranous expression of Glut-1 was regarded as positive. The expression of Glut-1 (the cut-off for positivity was determined as H-score 15) was found in 69/225 malignant melanomas. The number of positive cases and the H-score of Glut-1 increased where there was a higher Breslow thickness (p < 0.00001) when comparing pT1- pT4 MM groups. All benign nevi were classified as negative. In conclusion, the membranous expression of Glut-1 is a common feature of a malignant melanoma but this type of expression is very rare in benign melanocytic nevi. Our results suggest that the membranous expression of Glut-1 can be used as a surrogate marker in the assessing of the biological nature of benign and malignant melanocytic lesions. However, despite its high specificity, the sensitivity of this marker is relatively low. Moreover, due to the fact that the increased expression of Glut-1 correlates with a shorter survival period (10-year disease free survival, recurrence free survival and metastasis free survival and MFS), it can be used as a prognostically adverse factor.
Collapse
Affiliation(s)
- Miroslav Důra
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
- Department of Dermatology and Venereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kristýna Němejcová
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Radek Jakša
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Michaela Bártů
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Ondřej Kodet
- Department of Dermatology and Venereology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ivana Tichá
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Romana Michálková
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Studnickova 2, 12800, Prague 2, Czech Republic.
| |
Collapse
|
277
|
Shehata HM, Murphy AJ, Lee MKS, Gardiner CM, Crowe SM, Sanjabi S, Finlay DK, Palmer CS. Sugar or Fat?-Metabolic Requirements for Immunity to Viral Infections. Front Immunol 2017; 8:1311. [PMID: 29085369 PMCID: PMC5649203 DOI: 10.3389/fimmu.2017.01311] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/28/2017] [Indexed: 12/20/2022] Open
Abstract
The realization that an intricate link exists between the metabolic state of immune cells and the nature of the elicited immune responses has brought a dramatic evolution to the field of immunology. We will focus on how metabolic reprogramming through the use of glycolysis and fatty-acid oxidation (sugar or fat) regulates the capacity of immune cells to mount robust and effective immune responses. We will also discuss how fine-tuning sugar and fat metabolism may be exploited as a novel immunotherapeutic strategy to fight viral infections or improve vaccine efficacy.
Collapse
Affiliation(s)
- Hesham M Shehata
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, United States
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Man Kit Sam Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Clair M Gardiner
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Suzanne M Crowe
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Shomyseh Sanjabi
- Virology and Immunology, Gladstone Institutes, San Francisco, CA, United States
| | - David K Finlay
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Clovis Steve Palmer
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia.,Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
278
|
Metabolic pathway for the universal fluorescent recognition of tumor cells. Oncotarget 2017; 8:76108-76115. [PMID: 29100296 PMCID: PMC5652690 DOI: 10.18632/oncotarget.18551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/05/2017] [Indexed: 01/27/2023] Open
Abstract
Quantification of circulating tumor cells (CTCs) in blood samples from cancer patients is a non-invasive approach to monitoring the status of the disease. Most of the methods proposed in the recent years are phenomenological and rely on the use of antibodies labelled with fluorophores, magnetic particles, or immobilized on surfaces to capture the CTCs. Herein, we designed and optimized a method that employs a glucose analogue labelled with a fluorophore which takes advantage of the different metabolic pathways of cancer cells to discern them from normal ones. Notably, we demonstrate that fluorescence signal in tumor cells can be greatly maximized by applying hyperoxia conditions without damaging the cells. These results are demonstrated by means of confocal fluorescence and flow-cytometry measurements in peripheral blood mononuclear cells (PBMC) extracted after Ficoll of human blood samples and spiked with a known concentration of MCF-7 tumor cells.
Collapse
|
279
|
Luengo A, Gui DY, Vander Heiden MG. Targeting Metabolism for Cancer Therapy. Cell Chem Biol 2017; 24:1161-1180. [PMID: 28938091 PMCID: PMC5744685 DOI: 10.1016/j.chembiol.2017.08.028] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/06/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
Metabolic reprogramming contributes to tumor development and introduces metabolic liabilities that can be exploited to treat cancer. Chemotherapies targeting metabolism have been effective cancer treatments for decades, and the success of these therapies demonstrates that a therapeutic window exists to target malignant metabolism. New insights into the differential metabolic dependencies of tumors have provided novel therapeutic strategies to exploit altered metabolism, some of which are being evaluated in preclinical models or clinical trials. Here, we review our current understanding of cancer metabolism and discuss how this might guide treatments targeting the metabolic requirements of tumor cells.
Collapse
Affiliation(s)
- Alba Luengo
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dan Y Gui
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
280
|
El Hilali M, Reux B, Debiton E, Leal F, Galmier MJ, Vivier M, Chezal JM, Miot-Noirault E, Coudert P, Weber V. Linker structure-activity relationships in fluorodeoxyglucose chlorambucil conjugates for tumor-targeted chemotherapy. Bioorg Med Chem 2017; 25:5692-5708. [PMID: 28927903 DOI: 10.1016/j.bmc.2017.08.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/28/2017] [Accepted: 08/25/2017] [Indexed: 01/18/2023]
Abstract
Nitrogen mustards, such as chlorambucil (CLB), can cause adverse side-effects due to ubiquitous distribution in non-target organs. To minimize this toxicity, strategies of tumor-targeting drug delivery have been developed, where a cytotoxic warhead is linked to a tumor-cell-specific small ligand. Malignant cells exhibit marked glucose avidity and an accelerated metabolism by aerobic glycolysis, known as the Warburg effect, and recognized as a hallmark of cancer. A targeting approach exploiting the Warburg effect by conjugation of CLB to 2-fluoro-2-deoxyglucose (FDG) was previously reported and identified two peracetylated glucoconjugates 2 and 3 with promising antitumor activities in vivo. These results prompted us to investigate the importance of the spacer in this tumor-targeting glucose-based conjugates. Here we report the chemical synthesis and an in vitro cytotoxicity evaluation, using a 5-member panel of human tumor cell lines and human fibroblasts, of 16 new CLB glucoconjugates in which the alkylating drug is attached to the C-1 position of FDG via different linkages. We studied the structure-activity relationships in the linker, and evidenced the positive impact of an aromatic linker on in vitro cytotoxicity: compound 51 proved to be the most active FDG-CLB glucoside, characterized by a bis-aromatic spacer tethered to CLB through an amide function.
Collapse
Affiliation(s)
- Mostafa El Hilali
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Bastien Reux
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Eric Debiton
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Fernand Leal
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Marie-Josephe Galmier
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Magali Vivier
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Elisabeth Miot-Noirault
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Pascal Coudert
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France
| | - Valérie Weber
- Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
281
|
Shimada H, Kogure N, Noro E, Kudo M, Sugawara K, Sato I, Shimizu K, Kobayashi M, Suzuki D, Parvin R, Saito-Ito T, Uruno A, Saito-Hakoda A, Rainey WE, Ito S, Yokoyama A, Sugawara A. High glucose stimulates expression of aldosterone synthase ( CYP11B2) and secretion of aldosterone in human adrenal cells. FEBS Open Bio 2017; 7:1410-1421. [PMID: 28904869 PMCID: PMC5586344 DOI: 10.1002/2211-5463.12277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/11/2017] [Accepted: 07/26/2017] [Indexed: 11/09/2022] Open
Abstract
Aldosterone synthase is the key rate‐limiting enzyme in adrenal aldosterone production, and induction of its gene (CYP11B2) results in the progression of hypertension. As hypertension is a frequent complication among patients with diabetes, we set out to elucidate the link between diabetes mellitus and hypertension. We examined the effects of high glucose on CYP11B2 expression and aldosterone production using human adrenal H295R cells and a stable H295R cell line expressing a CYP11B2 5′‐flanking region/luciferase cDNA chimeric construct. d‐glucose (d‐glu), but not its enantiomer l‐glucose, dose dependently induced CYP11B2 transcription and mRNA expression. A high concentration (450 mg·dL−1) of d‐glu time dependently induced CYP11B2 transcription and mRNA expression. Moreover, high glucose stimulated secretion of aldosterone into the media. Transient transfection studies using deletion mutants/nerve growth factor‐induced clone B (NGFIB) response element 1 (NBRE‐1) point mutant of CYP11B2 5′‐flanking region revealed that the NBRE‐1 element, known to be activated by transcription factors NGFIB and NURR1, was responsible for the high glucose‐mediated effect. High glucose also induced the mRNA expression of these transcription factors, especially that of NURR1, but NURR1 knockdown using its siRNA did not affect high glucose‐induced CYP11B2 mRNA expression. Taken together, it is speculated that high glucose may induce CYP11B2 transcription via the NBRE‐1 element in its 5′‐flanking region, resulting in the increase in aldosterone production although high glucose‐induced NURR1 is not directly involved in the effect. Additionally, glucose metabolism and calcium channels were found to be involved in the high glucose effect. Our observations suggest one possible explanation for the high incidence of hypertension in patients with diabetes.
Collapse
Affiliation(s)
- Hiroki Shimada
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Naotaka Kogure
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Erika Noro
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Masataka Kudo
- Division of Nephrology, Endocrinology and Vascular Medicine Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Kaori Sugawara
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Ikuko Sato
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Kyoko Shimizu
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Makoto Kobayashi
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Dai Suzuki
- Department of Pediatrics Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Rehana Parvin
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Takako Saito-Ito
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Akira Uruno
- Department of Medical Biochemistry Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Akiko Saito-Hakoda
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - William E Rainey
- Department of Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor MI USA
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology and Vascular Medicine Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| | - Akira Sugawara
- Department of Molecular Endocrinology Tohoku University Graduate School of Medicine Sendai Miyagi Japan
| |
Collapse
|
282
|
Zielonka J, Sikora A, Hardy M, Ouari O, Vasquez-Vivar J, Cheng G, Lopez M, Kalyanaraman B. Mitochondria-Targeted Triphenylphosphonium-Based Compounds: Syntheses, Mechanisms of Action, and Therapeutic and Diagnostic Applications. Chem Rev 2017; 117:10043-10120. [PMID: 28654243 PMCID: PMC5611849 DOI: 10.1021/acs.chemrev.7b00042] [Citation(s) in RCA: 1035] [Impact Index Per Article: 129.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria are recognized as one of the most important targets for new drug design in cancer, cardiovascular, and neurological diseases. Currently, the most effective way to deliver drugs specifically to mitochondria is by covalent linking a lipophilic cation such as an alkyltriphenylphosphonium moiety to a pharmacophore of interest. Other delocalized lipophilic cations, such as rhodamine, natural and synthetic mitochondria-targeting peptides, and nanoparticle vehicles, have also been used for mitochondrial delivery of small molecules. Depending on the approach used, and the cell and mitochondrial membrane potentials, more than 1000-fold higher mitochondrial concentration can be achieved. Mitochondrial targeting has been developed to study mitochondrial physiology and dysfunction and the interaction between mitochondria and other subcellular organelles and for treatment of a variety of diseases such as neurodegeneration and cancer. In this Review, we discuss efforts to target small-molecule compounds to mitochondria for probing mitochondria function, as diagnostic tools and potential therapeutics. We describe the physicochemical basis for mitochondrial accumulation of lipophilic cations, synthetic chemistry strategies to target compounds to mitochondria, mitochondrial probes, and sensors, and examples of mitochondrial targeting of bioactive compounds. Finally, we review published attempts to apply mitochondria-targeted agents for the treatment of cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Lodz University of Technology, ul. Wroblewskiego 15, 93-590 Lodz, Poland
| | - Micael Hardy
- Aix Marseille Univ, CNRS, ICR, UMR 7273, 13013 Marseille, France
| | - Olivier Ouari
- Aix Marseille Univ, CNRS, ICR, UMR 7273, 13013 Marseille, France
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Gang Cheng
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| | - Marcos Lopez
- Translational Biomedical Research Group, Biotechnology Laboratories, Cardiovascular Foundation of Colombia, Carrera 5a No. 6-33, Floridablanca, Santander, Colombia, 681003
- Graduate Program of Biomedical Sciences, Faculty of Health, Universidad del Valle, Calle 4B No. 36-00, Cali, Colombia, 760032
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
- Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States
| |
Collapse
|
283
|
Arbe MF, Fondello C, Agnetti L, Álvarez GM, Tellado MN, Glikin GC, Finocchiaro LME, Villaverde MS. Inhibition of bioenergetic metabolism by the combination of metformin and 2-deoxyglucose highly decreases viability of feline mammary carcinoma cells. Res Vet Sci 2017; 114:461-468. [PMID: 28802138 DOI: 10.1016/j.rvsc.2017.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/19/2017] [Accepted: 07/31/2017] [Indexed: 01/05/2023]
Abstract
Feline mammary carcinoma (FMC) is a highly aggressive pathology that has been proposed as an interesting model of breast cancer disease, especially for the hormone refractory subgroup. Recently, cancer cell metabolism has been described as a hallmark of cancer cells. Here, we investigate the effects and mechanism of metabolic modulation by metformin (MET, anti-diabetic drug), 2-deoxyglucose (2DG, hexokinase inhibitor) or a combination of both drugs, MET/2DG on two established FMC cells lines: AlRB (HER2 (3+) and Ki67<5%) and AlRATN (HER2 (-) and Ki67>15%). We found that treatments significantly decreased both FMC cells viability by up to 80%. AlRB resulted more sensitive to 2DG than AlRATN (IC50: 3.15 vs 6.32mM, respectively). The combination of MET/2DG potentiated the effects of the individually added drugs on FMC cells. In addition, MET/2DG caused an increased in intracellular oxidants, autophagic vesicles and completely inhibited colony formation. Conversely, only MET significantly altered plasma membrane integrity, presented late apoptotic/necrotic cells and increased both glucose consumption and lactate concentration. Our results support further studies to investigate the potential use of this metabolic modulation approach in a clinical veterinary setting.
Collapse
Affiliation(s)
- María Florencia Arbe
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina
| | - Chiara Fondello
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina
| | - Lucrecia Agnetti
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina
| | - Gabriel Martín Álvarez
- Cátedra de Química Biológica, Facultad de Veterinaria, Universidad de Buenos, Ciudad Autónoma de Buenos Aires, Argentina
| | - Matías Nicolás Tellado
- Cátedra de Química Biológica, Facultad de Veterinaria, Universidad de Buenos, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gerardo Claudio Glikin
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina
| | - Liliana María Elena Finocchiaro
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina
| | - Marcela Solange Villaverde
- Unidad de Transferencia Genética, Instituto de Oncología Dr. Ángel Roffo, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma Buenos Aires, Argentina.
| |
Collapse
|
284
|
Guerra F, Arbini AA, Moro L. Mitochondria and cancer chemoresistance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:686-699. [DOI: 10.1016/j.bbabio.2017.01.012] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/07/2023]
|
285
|
Xu L, Li H, Wu L, Huang S. YBX1 promotes tumor growth by elevating glycolysis in human bladder cancer. Oncotarget 2017; 8:65946-65956. [PMID: 29029484 PMCID: PMC5630384 DOI: 10.18632/oncotarget.19583] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022] Open
Abstract
Aerobic glycolysis, also known as Warburg effect, is a key hallmark of cancers. The Y-box-binding protein 1 (YBX1) is a well-known oncoprotein implicated in multiple malignant phenotypes of cancers. Meanwhile, little is known about the oncogenic functions and mechanisms of YBX1 in bladder cancer. Based on gene set enrichment analysis (GSEA) of TCGA RNAseq data, we find that YBX1 was profoundly involved in the glycolysis part of glucose metabolism. Loss- and gain-of-function studies show that YBX1 can enhance glycolysis as revealed by expression of glycolytic enzymes, glucose uptake, lactate secretion and extracellular acidification rate (ECAR). Inhibition of glycolysis completely compromises the tumor-promoting effect of YBX1 on tumor growth. Mechanistically, YBX1 regulates the expression of c-Myc and HIF1α, which further upregulate glycolytic enzymes to facilitate glycolysis. Moreover, in vivo study further confirms that genetic silencing of YBX1 markedly attenuates tumor growth and this tumor-suppressive effect is largely dependent on reduced glycolysis. Taken together, these results, as a proof of principle, provide a novel insight into the oncogenic role of YBX1 in glycolysis and suggest the potential therapeutic strategy by targeting YBX1 in bladder cancer.
Collapse
Affiliation(s)
- Liuyu Xu
- Department of Urology, QianFoShan Hospital Affiliated to Shandong University, Jinan 250014, P. R. China
| | - Hongyun Li
- Department of Urology, QianFoShan Hospital Affiliated to Shandong University, Jinan 250014, P. R. China
| | - Longchao Wu
- Department of Urology, Penglai People's Hospital of Shandong, Penglai 265600, P. R. China
| | - Shiming Huang
- Department of Urology, QianFoShan Hospital Affiliated to Shandong University, Jinan 250014, P. R. China
| |
Collapse
|
286
|
Hirschhorn T, Levi-Hofman M, Danziger O, Smorodinsky NI, Ehrlich M. Differential molecular regulation of processing and membrane expression of Type-I BMP receptors: implications for signaling. Cell Mol Life Sci 2017; 74:2645-2662. [PMID: 28357470 PMCID: PMC11107780 DOI: 10.1007/s00018-017-2488-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
The Type-I bone morphogenetic protein receptors (BMPRs), BMPR1A and BMPR1B, present the highest sequence homology among BMPRs, suggestive of functional similitude. However, sequence elements within their extracellular domain, such as signal sequence or N-glycosylation motifs, may result in differential regulation of biosynthetic processing and trafficking and in alterations to receptor function. We show that (i) BMPR1A and the ubiquitous isoform of BMPR1B differed in mode of translocation into the endoplasmic reticulum; and (ii) BMPR1A was N-glycosylated while BMPR1B was not, resulting in greater efficiency of processing and plasma membrane expression of BMPR1A. We further demonstrated the importance of BMPR1A expression and glycosylation in ES-2 ovarian cancer cells, where (i) CRISPR/Cas9-mediated knockout of BMPR1A abrogated BMP2-induced Smad1/5/8 phosphorylation and reduced proliferation of ES-2 cells and (ii) inhibition of N-glycosylation by site-directed mutagenesis, or by tunicamycin or 2-deoxy-D-glucose treatments, reduced biosynthetic processing and plasma membrane expression of BMPR1A and BMP2-induced Smad1/5/8 phosphorylation.
Collapse
Affiliation(s)
- Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Levi-Hofman
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Oded Danziger
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nechama I Smorodinsky
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
287
|
Sun L, Yin Y, Clark LH, Sun W, Sullivan SA, Tran AQ, Han J, Zhang L, Guo H, Madugu E, Pan T, Jackson AL, Kilgore J, Jones HM, Gilliam TP, Zhou C, Bae-Jump VL. Dual inhibition of glycolysis and glutaminolysis as a therapeutic strategy in the treatment of ovarian cancer. Oncotarget 2017; 8:63551-63561. [PMID: 28969010 PMCID: PMC5609942 DOI: 10.18632/oncotarget.18854] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/04/2017] [Indexed: 01/12/2023] Open
Abstract
Cancer cell metabolism is required to support the biosynthetic demands of cell growth and cell division, and to maintain reduction oxidaton (redox) homeostasis. This study was designed to test the effects of glucose and glutamine on ovarian cancer cell growth and explore the inter-relationship between glycolysis and glutaminolysis. The SKOV3, IGROV-1 and Hey ovarian cancer cell lines were assayed for glucose, pyruvate and glutamine dependence by analyzing cytotoxicity, cell cycle progression, apoptosis and ATP production. As determined by MTT assay, glucose stimulated cell growth while the combination of glucose, glutamine and pyruvate resulted in the greatest stimulation of cell proliferation. Furthermore, 2-deoxy-glucose (2-DG) and 3-bromopyruvate (3-BP) induced apoptosis, caused G1 phase cell cycle arrest and reduced glycolytic activity. Moreover, 2-DG in combination with a low dose of aminooxyacetate (AOA) synergistically increased the sensitivity to 2-DG in the inhibition of cell growth in the ovarian cancer cell lines. These studies suggest that dual inhibition of glycolysis and glutaminolysis may be a promising therapeutic strategy for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Li Sun
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yajie Yin
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China.,School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leslie H Clark
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie A Sullivan
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Arthur-Quan Tran
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianjun Han
- Department of Surgical Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China
| | - Lu Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China
| | - Hui Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China
| | - Esther Madugu
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tommy Pan
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda L Jackson
- Division of Gynecologic Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua Kilgore
- Houston Methodist Gynecologic Oncology Associates, Houston, TX, USA
| | - Hannah M Jones
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Timothy P Gilliam
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
288
|
Suvarna S, Das U, KC S, Mishra S, Sudarshan M, Saha KD, Dey S, Chakraborty A, Narayana Y. Synthesis of a novel glucose capped gold nanoparticle as a better theranostic candidate. PLoS One 2017; 12:e0178202. [PMID: 28582426 PMCID: PMC5459428 DOI: 10.1371/journal.pone.0178202] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 05/09/2017] [Indexed: 01/17/2023] Open
Abstract
Gold nanoparticles are predominantly used in diagnostics, therapeutics and biomedical applications. The present study has been designed to synthesize differently capped gold nanoparticles (AuNps) by a simple, one-step, room temperature procedure and to evaluate the potential of these AuNps for biomedical applications. The AuNps are capped with glucose, 2-deoxy-D-glucose (2DG) and citrate using different reducing agents. This is the first report of synthesis of 2DG-AuNp by the simple room temperature method. The synthesized gold nanoparticles are characterized with UV-Visible Spectroscopy, Fourier transform infrared spectroscopy (FTIR), Transmission electron microscopy (TEM) and selected area electron diffraction (SAED), Dynamic light scattering (DLS), and Energy-dispersive X-ray spectroscopy (SEM-EDS). Surface-enhanced Raman scattering (SERS) study of the synthesized AuNps shows increase in Raman signals up to 50 times using 2DG. 3-(4, 5-dimethylthiozol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay has been performed using all the three differently capped AuNps in different cell lines to assess cytotoxcity if any, of the nanoparticles. The study shows that 2DG-AuNps is a better candidate for theranostic application.
Collapse
Affiliation(s)
- Saritha Suvarna
- Department of Studies in Physics, Mangalore University, Mangalagangotri, Karnataka, India
| | - Ujjal Das
- Department of Physiology, Centre for Nanoscience and Nanotechnology and Centre with Potential for Excellence in Particular Area (CPEPA), University of Calcutta, Kolkata, West Bengal, India
| | - Sunil KC
- Department of Studies in Physics, Mangalore University, Mangalagangotri, Karnataka, India
| | - Snehasis Mishra
- Cancer Biology & Inflammatory Disorder Division, IICB, Kolkata, West Bengal, India
| | - Mathummal Sudarshan
- UGC-DAE Consortium for Scientific Research Centre, Kolkata, West Bengal, India
| | - Krishna Das Saha
- Cancer Biology & Inflammatory Disorder Division, IICB, Kolkata, West Bengal, India
| | - Sanjit Dey
- Department of Physiology, Centre for Nanoscience and Nanotechnology and Centre with Potential for Excellence in Particular Area (CPEPA), University of Calcutta, Kolkata, West Bengal, India
| | | | - Y. Narayana
- Department of Studies in Physics, Mangalore University, Mangalagangotri, Karnataka, India
- * E-mail:
| |
Collapse
|
289
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. Differentiation of human induced pluripotent stem cells in William's E initiation medium supplemented with 3‑bromopyruvate and 2‑deoxy‑d‑glucose. Mol Med Rep 2017; 15:3719-3723. [PMID: 28440498 DOI: 10.3892/mmr.2017.6474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 02/21/2017] [Indexed: 11/05/2022] Open
Abstract
Hepatocyte selection medium (HSM) is deprived of glucose and supplemented with galactose, and is based on Leibovitz's‑15 (L15) medium. HSM may promote the differentiation of human induced pluripotent stem (iPS) cells towards hepatocyte lineage. These culture conditions result in increased expression of galactokinase (GALK)‑1 and GALK2. However, iPS cells do not survive in HSM. Two potential alternatives to glucose deprivation are treatment with 3‑bromopyruvate (3BP), an analogue of pyruvate, and 2‑deoxy‑d‑glucose (2DG), an analogue of glucose. The promoters of GALK1 and GALK2 were subcloned using the pMetLuc2 reporter plasmid to make pMetLuc2/GALK1 and pMetLuc2/GALK2, respectively. 201B7 human iPS cells were transfected with the reporter plasmids, cultured in HSM and analyzed by luciferase assay. Furthermore, 201B7 cells were cultured in L15, William's E (WE) or Dulbecco's modified Eagle's medium/nutrient mixture F‑12 Ham (DF12) supplemented with 3BP, 2DG or a combination of the two, for 15 days, and subjected to reverse transcription‑quantitative polymerase chain reaction to measure the levels of α‑fetoprotein (AFP) mRNA expression. Metridia luciferase activity was significantly higher in cells cultured in HSM compared with those in ReproFF medium (P<0.05). 3BP and 2DG treatment, alone or in combination, decreased AFP expression levels in cells cultured in L15 and DF12. The combination of 3BP+2DG increased the expression levels of AFP in WE. Without 3BP or 2DG, AFP expression was higher in L15 compared with WE or DF12. The promoters of GALK1 and GALK2 were activated in 201B7 cells cultured in HSM, enabling survival using galactose as an energy source. 3BP and 2DG supplementation in WE medium may promote the differentiation of iPS cells to the hepatocyte lineage.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| |
Collapse
|
290
|
Metabolic Portraits of Breast Cancer by HR MAS MR Spectroscopy of Intact Tissue Samples. Metabolites 2017; 7:metabo7020018. [PMID: 28509845 PMCID: PMC5487989 DOI: 10.3390/metabo7020018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/20/2017] [Accepted: 05/09/2017] [Indexed: 12/17/2022] Open
Abstract
Despite progress in early detection and therapeutic strategies, breast cancer remains the second leading cause of cancer-related death among women globally. Due to the heterogeneity and complexity of tumor biology, breast cancer patients with similar diagnosis might have different prognosis and response to treatment. Thus, deeper understanding of individual tumor properties is necessary. Cancer cells must be able to convert nutrients to biomass while maintaining energy production, which requires reprogramming of central metabolic processes in the cells. This phenomenon is increasingly recognized as a potential target for treatment, but also as a source for biomarkers that can be used for prognosis, risk stratification and therapy monitoring. Magnetic resonance (MR) metabolomics is a widely used approach in translational research, aiming to identify clinically relevant metabolic biomarkers or generate novel understanding of the molecular biology in tumors. Ex vivo proton high-resolution magic angle spinning (HR MAS) MR spectroscopy is widely used to study central metabolic processes in a non-destructive manner. Here we review the current status for HR MAS MR spectroscopy findings in breast cancer in relation to glucose, amino acid and choline metabolism.
Collapse
|
291
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. 2‑Deoxy‑D‑glucose initiates hepatocyte differentiation in human induced pluripotent stem cells. Mol Med Rep 2017; 15:3083-3087. [PMID: 28358426 DOI: 10.3892/mmr.2017.6405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 02/15/2017] [Indexed: 11/06/2022] Open
Abstract
To initiate hepatocyte differentiation in human induced pluripotent stem (iPS) cells, cells are cultured in a medium lacking glucose but supplemented with galactose (hepatocyte selection medium, HSM) or in medium supplemented with oncostatin M and small molecules (hepatocyte differentiation inducer, HDI). In the present study, 2‑Deoxy‑D‑glucose (2DG), an analogue of glucose, was utilized instead of glucose deprivation and the effect of 2DG supplementation on iPS differentiation was examined. First, 201B7 cells, an iPS cell line, were cultured in HSM or HDI media for 2 days and then subjected to reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) in order to analyze expression levels of established hepatocyte markers, including cytosolic aspartate aminotransferase (AST), mitochondrial AST, alanine aminotransferase (ALT), and glycerol kinase. mRNA expression levels of mitochondrial AST, ALT, and glycogen synthase significantly increased following culture in HSM and HDI compared with ReproFF media. Cytosolic AST mRNA expression levels significantly increased following culture in HDI compared with ReproFF media, but not in HSM. To test the effect of 2DG on iPS differentiation, 201B7 cells were cultured in ReproFF, a feeder‑free medium that retains pluripotency, supplemented with 2DG. Following 7 days of culture, the cells were subjected to RT‑qPCR to analyze expression levels of α‑fetoprotein (AFP), a marker of immature hepatocytes. AFP mRNA expression levels significantly increased with the addition of 0.1 µM 2DG in the media, and galactose addition acted synergistically with 2DG to further upregulate AFP expression. In conclusion, the present study demonstrated that hepatocyte differentiation was initiated in iPS cells cultured in HSM and HDI media and that 2DG could be used as a supplement instead of glucose deprivation to initiate hepatocyte differentiation in iPS cells.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284‑0003, Japan
| |
Collapse
|
292
|
Magistroni R, Boletta A. Defective glycolysis and the use of 2-deoxy-D-glucose in polycystic kidney disease: from animal models to humans. J Nephrol 2017; 30:511-519. [PMID: 28390001 DOI: 10.1007/s40620-017-0395-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited renal disease characterized by bilateral renal cyst formation. ADPKD is one of the most common rare disorders, accounting for ~10% of all patients with end-stage renal disease (ESRD). ADPKD is a chronic disorder in which the gradual expansion of cysts that form in a minority of nephrons eventually causes loss of renal function due to the compression and degeneration of the surrounding normal parenchyma. Numerous deranged pathways have been identified in the cyst-lining epithelia, prompting the design of potential therapies. Several of these potential treatments have proved effective in slowing down disease progression in pre-clinical animal studies, while only one has subsequently been proven to effectively slow down disease progression in patients, and it has recently been approved for therapy in Europe, Canada and Japan. Among the affected cellular functions and pathways, recent investigations have described metabolic derangement in ADPKD as a major trait offering additional opportunities for targeted therapies. In particular, increased aerobic glycolysis (the Warburg effect) has been described as a prominent feature of ADPKD kidneys and its inhibition using the glucose analogue 2-deoxy-D-glucose (2DG) proved effective in slowing down disease progression in preclinical models of the disease. At the same time, previous clinical experiences have been reported with 2DG, showing that this compound is well tolerated in humans with minimal and reversible side effects. In this work, we review the literature and speculate that 2DG could be a good candidate for a clinical trial in humans affected by ADPKD.
Collapse
Affiliation(s)
- Riccardo Magistroni
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina, 58, 20132, Milan, Italy
- Division of Nephrology and Hypertension, San Raffaele Hospital, Milan, Italy
- Division of Nephrology and Dialysis, AOU Policlinico di Modena, Università di Modena e Reggio Emilia, Modena, Italy
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Via Olgettina, 58, 20132, Milan, Italy.
| |
Collapse
|
293
|
Targeting MYC Dependence by Metabolic Inhibitors in Cancer. Genes (Basel) 2017; 8:genes8040114. [PMID: 28362357 PMCID: PMC5406861 DOI: 10.3390/genes8040114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
MYC is a critical growth regulatory gene that is commonly overexpressed in a wide range of cancers. Therapeutic targeting of MYC transcriptional activity has long been a goal, but it has been difficult to achieve with drugs that directly block its DNA-binding ability. Additional approaches that exploit oncogene addiction are promising strategies against MYC-driven cancers. Also, drugs that target metabolic regulatory pathways and enzymes have potential for indirectly reducing MYC levels. Glucose metabolism and oxidative phosphorylation, which can be targeted by multiple agents, promote cell growth and MYC expression. Likewise, modulation of the signaling pathways and protein synthesis regulated by adenosine monophosphate-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) can also be an effective route for suppressing MYC translation. Furthermore, recent data suggest that metabolism of nucleotides, fatty acids and glutamine are exploited to alter MYC levels. Combination therapies offer potential new approaches to overcome metabolic plasticity caused by single agents. Although potential toxicities must be carefully controlled, new inhibitors currently being tested in clinical trials offer significant promise. Therefore, as both a downstream target of metabolism and an upstream regulator, MYC is a prominent central regulator of cancer metabolism. Exploiting metabolic vulnerabilities of MYC-driven cancers is an emerging research area with translational potential.
Collapse
|
294
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. Proliferation of sphere-forming hepatocellular carcinoma cells is suppressed in a medium without glucose and arginine, but with galactose and ornithine. Oncol Lett 2017; 13:1264-1268. [PMID: 28454244 PMCID: PMC5403635 DOI: 10.3892/ol.2017.5565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 08/25/2016] [Indexed: 01/04/2023] Open
Abstract
Resistance to sorafenib in hepatocellular carcinoma (HCC) cells exhibiting stemness was evaluated using a sphere formation assay. A hepatocyte selection medium (HSM) deficient in glucose and arginine was used to suppress the proliferation of cell spheres composed of HLF and PLC/PRF/5 HCC cells, which were subjected to a sphere formation assay. Cell spheres were cultured with sorafenib and subjected to a cell proliferation assay and the expression levels of cytochrome P450 (CYP3A4) were analyzed in RNA extracted from sphere-forming cells using reverse transcription-quantitative polymerase chain reaction. Sphere-forming PLC/PRF/5 cells were more resistant to sorafenib, as compared with control cells, exhibiting higher expression levels of CYP3A4. When cultured in HSM, suppressed proliferation was observed in the sphere-forming PLC/PRF/5 cells and in the control cells, with no significant variation between them. The results suggest that deprivation of glucose and arginine is a potential novel treatment for HCC.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
295
|
Zhao H, Duan Q, Zhang Z, Li H, Wu H, Shen Q, Wang C, Yin T. Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. J Cell Mol Med 2017; 21:2055-2067. [PMID: 28244691 PMCID: PMC5571518 DOI: 10.1111/jcmm.13126] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/01/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) and epithelial–mesenchymal transition (EMT)‐type cells are considered as underlying causes of chemoresistance, tumour recurrence and metastasis in pancreatic cancer. We aimed to describe the mechanisms – particularly glycolysis – involved in the regulation of the CSC and EMT phenotypes. We used a gemcitabine‐resistant (GR) Patu8988 cell line, which exhibited clear CSC and EMT phenotypes and showed reliance on glycolysis. Inhibition of glycolysis using 2‐deoxy‐D‐glucose (2‐DG) significantly enhanced the cytotoxicity of gemcitabine and inhibited the CSC and EMT phenotypes in GR cells both in vitro and in vivo. Intriguingly, the use of the reactive oxygen species (ROS) scavenger N‐acetylcysteine (NAC) restored the CSC and EMT phenotypes. H2O2 produced changes similar to those of 2‐DG, indicating that ROS were involved in the acquired cancer stemness and EMT phenotypes of GR cells. Moreover, doublecortin‐like kinase 1 (DCLK1), a pancreatic CSC marker, was highly expressed and regulated the stemness and EMT phenotypes in GR cell. Both 2‐DG and H2O2 treatment suppressed DCLK1 expression, which was also rescued by NAC. Together, these findings revealed that glycolysis promotes the expression of DCLK1 and maintains the CSC and EMT phenotypes via maintenance of low ROS levels in chemoresistant GR cells. The glycolysis‐ROS‐DCLK1 pathway may be potential targets for reversing the malignant behaviour of pancreatic cancer.
Collapse
Affiliation(s)
- Hengqiang Zhao
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingke Duan
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengle Zhang
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hehe Li
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heshui Wu
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Shen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chunyou Wang
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Yin
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
296
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. 2-Deoxyglucose and sorafenib synergistically suppress the proliferation and motility of hepatocellular carcinoma cells. Oncol Lett 2017; 13:800-804. [PMID: 28356961 PMCID: PMC5351389 DOI: 10.3892/ol.2016.5510] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/18/2016] [Indexed: 12/25/2022] Open
Abstract
Cancer cells consume more glucose than normal cells, mainly due to their increased rate of glycolysis. 2-Deoxy-d-glucose (2DG) is an analogue of glucose, and sorafenib is a kinase inhibitor and molecular agent used to treat hepatocellular carcinoma (HCC). The present study aimed to demonstrate whether combining 2DG and sorafenib suppresses tumor cell proliferation and motility more effectively than either drug alone. HLF and PLC/PRF/5 HCC cells were incubated with sorafenib with or without 1 µM 2DG, and subjected to a proliferation assay. A scratch assay was then performed to analyze cell motility following the addition of 2DG and sorafenib in combination, and each agent alone. RNA was isolated and subjected to reverse transcription-quantitative polymerase chain reaction to analyze the expression of cyclin D1 and matrix metalloproteinase-9 (MMP9) following the addition of 2DG and sorafenib in combination and each agent alone. Proliferation was markedly suppressed in cells cultured with 1 µM 2DG and 30 µM sorafenib compared with cells cultured with either agent alone (P<0.05). In addition, levels of Cyclin D1 expression decreased in cells exposed to 3 µM sorafenib and 1 µM 2DG compared with cells exposed to 2DG or sorafenib alone (P<0.05). Scratch assay demonstrated that the distance between the growing edge of the cell sheet and the scratched line was shorter in cells cultured with sorafenib and 2DG than in cells cultured with 2DG or sorafenib alone (P<0.05). Levels of MMP9 expression decreased more in cells treated with both sorafenib and 2DG than in cells treated with 2DG or sorafenib alone (P<0.05). Therefore, 2DG and sorafenib in combination suppressed the proliferation and motility of HCC cells more effectively than 2DG or sorafenib alone, and a cancer treatment combining both drugs may be more effective than sorafenib alone.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
297
|
Araujo L, Khim P, Mkhikian H, Mortales CL, Demetriou M. Glycolysis and glutaminolysis cooperatively control T cell function by limiting metabolite supply to N-glycosylation. eLife 2017; 6. [PMID: 28059703 PMCID: PMC5257256 DOI: 10.7554/elife.21330] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022] Open
Abstract
Rapidly proliferating cells switch from oxidative phosphorylation to aerobic glycolysis plus glutaminolysis, markedly increasing glucose and glutamine catabolism. Although Otto Warburg first described aerobic glycolysis in cancer cells >90 years ago, the primary purpose of this metabolic switch remains controversial. The hexosamine biosynthetic pathway requires glucose and glutamine for de novo synthesis of UDP-GlcNAc, a sugar-nucleotide that inhibits receptor endocytosis and signaling by promoting N-acetylglucosamine branching of Asn (N)-linked glycans. Here, we report that aerobic glycolysis and glutaminolysis co-operatively reduce UDP-GlcNAc biosynthesis and N-glycan branching in mouse T cell blasts by starving the hexosamine pathway of glucose and glutamine. This drives growth and pro-inflammatory TH17 over anti-inflammatory-induced T regulatory (iTreg) differentiation, the latter by promoting endocytic loss of IL-2 receptor-α (CD25). Thus, a primary function of aerobic glycolysis and glutaminolysis is to co-operatively limit metabolite supply to N-glycan biosynthesis, an activity with widespread implications for autoimmunity and cancer. DOI:http://dx.doi.org/10.7554/eLife.21330.001
Collapse
Affiliation(s)
- Lindsey Araujo
- Department of Microbiology and Molecular Genetics, University of California, Irvine, United States
| | - Phillip Khim
- Department of Neurology and Institute for Immunology, University of California, Irvine, United States
| | - Haik Mkhikian
- Department of Pathology and Laboratory Medicine, University of California, Irvine, United States
| | - Christie-Lynn Mortales
- Department of Microbiology and Molecular Genetics, University of California, Irvine, United States
| | - Michael Demetriou
- Department of Neurology and Institute for Immunology, University of California, Irvine, United States
| |
Collapse
|
298
|
Sun L, Liu X, Fu H, Zhou W, Zhong D. 2-Deoxyglucose Suppresses ERK Phosphorylation in LKB1 and Ras Wild-Type Non-Small Cell Lung Cancer Cells. PLoS One 2016; 11:e0168793. [PMID: 28033353 PMCID: PMC5198974 DOI: 10.1371/journal.pone.0168793] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/06/2016] [Indexed: 12/31/2022] Open
Abstract
Tumor cells rely on aerobic glycolysis to generate ATP, namely the "Warburg" effect. 2-deoxyglucose (2-DG) is well characterized as a glycolytic inhibitor, but its effect on cellular signaling pathways has not been fully elucidated. Herein, we sought to investigate the effect of 2-DG on ERK function in lung cancer cells. We found that 2-DG inhibits ERK phosphorylation in a time and dose-dependent manner in lung cancer cells. This inhibition requires functional LKB1. LKB1 knockdown in LKB1 wildtype cells correlated with an increase in the basal level of p-ERK. Restoration of LKB1 in LKB1-null cells significantly inhibits ERK activation. Blocking AMPK function with AMPK inhibitor, AMPK siRNA or DN-AMPK diminishes the inhibitory effect of 2-DG on ERK, suggesting that 2-DG—induced ERK inhibition is mediated by LKB1/AMPK signaling. Moreover, IGF1-induced ERK phosphorylation is significantly decreased by 2-DG. Conversely, a subset of oncogenic mutants of K-Ras, the main upstream regulator of ERK, blocks 2-DG—induced LKB1/AMPK signaling. These findings reveal the potential cross-talk between LKB1/AMPK and ERK signaling and help to better understand the mechanism of action of 2-DG.
Collapse
Affiliation(s)
- Linlin Sun
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Xiuju Liu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine and the Department of Human Genetics Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (DZ); (WZ)
| | - Diansheng Zhong
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, P.R. China
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, P.R. China
- * E-mail: (DZ); (WZ)
| |
Collapse
|
299
|
Němejcová K, Rosmusová J, Bártů M, Důra M, Tichá I, Dundr P. Expression of Glut-1 in Normal Endometrium and Endometrial Lesions: Analysis of 336 Cases. Int J Surg Pathol 2016; 25:389-396. [DOI: 10.1177/1066896916683510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Glucose transporter-1 (Glut-1) is a membrane glycoprotein that is, together with other glucose transporters, responsible for the regulation of glucose uptake. An increased expression of this protein seems to be a general feature of several malignant tumors that are able to reprogram their metabolism and switch from oxidative phosphorylation to aerobic glycolysis. Methods: We performed comprehensive immunohistochemical analysis of Glut-1 expression in 336 endometrial samples, including tumors, nontumor lesions, and normal tissues. Results: Expression of Glut-1 was found in 87% of endometrioid carcinomas (160/184 cases), 100% of serous carcinomas (29/29 cases), 100% of clear cell carcinomas (17/17 cases), 50% of polyps with atypical hyperplasia (8/16 cases), 12.5% of polyps with non-atypical hyperplasia (3/24 cases), 77% of hyperplasias with atypias (10/13 cases), 9% of hyperplasias without atypias (1/11 cases), 87% of secretory endometrium samples (13/15 cases), and in none of the nonsecretory endometrium samples (0/27 cases). In endometrioid carcinomas, Glut-1 was expressed in a marked geographical pattern. In nontumor lesions, its expression was more common in atypical hyperplasia and polyps with atypical hyperplasia compared with polyps with non-atypical hyperplasia and hyperplasias without atypia ( P = .00032). Conclusion: Our study confirms the high expression of Glut-1 not only in endometrioid carcinomas but also in other carcinomas of endometrium including clear cell and serous types. Glut-1 expression can be used as a surrogate marker in differential diagnosis between hyperplasia with and without atypia. Because of common Glut-1 expression in malignant tumors, therapeutic strategies influencing this protein or its signaling pathways can be beneficial.
Collapse
Affiliation(s)
- Kristýna Němejcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jana Rosmusová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Michaela Bártů
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Miroslav Důra
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Dermatovenerology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ivana Tichá
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Dundr
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
300
|
Ranftler C, Meisslitzer-Ruppitsch C, Neumüller J, Ellinger A, Pavelka M. Golgi apparatus dis- and reorganizations studied with the aid of 2-deoxy-D-glucose and visualized by 3D-electron tomography. Histochem Cell Biol 2016; 147:415-438. [PMID: 27975144 PMCID: PMC5359389 DOI: 10.1007/s00418-016-1515-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2016] [Indexed: 12/31/2022]
Abstract
We studied Golgi apparatus disorganizations and reorganizations in human HepG2 hepatoblastoma cells by using the nonmetabolizable glucose analogue 2-deoxy-d-glucose (2DG) and analyzing the changes in Golgi stack architectures by 3D-electron tomography. Golgi stacks remodel in response to 2DG-treatment and are replaced by tubulo-glomerular Golgi bodies, from which mini-Golgi stacks emerge again after removal of 2DG. The Golgi stack changes correlate with the measured ATP-values. Our findings indicate that the classic Golgi stack architecture is impeded, while cells are under the influence of 2DG at constantly low ATP-levels, but the Golgi apparatus is maintained in forms of the Golgi bodies and Golgi stacks can be rebuilt as soon as 2DG is removed. The 3D-electron microscopic results highlight connecting regions that interlink membrane compartments in all phases of Golgi stack reorganizations and show that the compact Golgi bodies mainly consist of continuous intertwined tubules. Connections and continuities point to possible new transport pathways that could substitute for other modes of traffic. The changing architectures visualized in this work reflect Golgi stack dynamics that may be essential for basic cell physiologic and pathologic processes and help to learn, how cells respond to conditions of stress.
Collapse
Affiliation(s)
- Carmen Ranftler
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | | | - Josef Neumüller
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Adolf Ellinger
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Margit Pavelka
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria.
| |
Collapse
|