1
|
Maloba GO, Were T, Barasa E, Mohamed N, Arshi A, Gallyas F. Synergistic Effects of 2-Deoxyglucose and Diclofenac Sodium on Breast Cancer Cells: A Comparative Evaluation of MDA-231 and MCF7 Cells. Int J Mol Sci 2025; 26:4894. [PMID: 40430033 PMCID: PMC12112485 DOI: 10.3390/ijms26104894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Resistance of breast cancers to chemotherapy remains a global challenge to date. Drug combination studies between anti-cancer agents are increasingly becoming therapeutic strategies, geared towards alleviating breast cancers. Previously, 2-deoxyglucose has been shown to target and interrupt glycolysis. Available evidence also suggests that diclofenac, which was originally designed as a pain reliever, could inhibit the proliferation of breast cancer cells. However, the reverse Warburg effect and other metabolic reprogramming mechanisms in breast cancers limit the pharmacological application of both 2-deoxyglucose and diclofenac as mono-therapeutic agents. The present study explores the additive anti-cancer effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells. In this study, MDA-231 and MCF7 cells were treated with 2-deoxyglucose and diclofenac sodium in single and combination doses before being evaluated for viability, cell growth, reactive oxygen species, apoptotic and necrotic phases, and migration abilities. Additionally, immunoblotting of pro-apoptotic proteins, Caspase-3 and Caspase-9, and a hypoxia-inducible factor-1 alpha, was also performed. The results showed that combination treatments of the cells with the drugs exhibited additive anti-cancer effects by limiting proliferation, enhancing cytotoxic reactive oxygen species generation, enhancing apoptosis and necrosis, limiting colony formation and expansion of cells, and inhibiting cell migration. The degrees of cytotoxicity of combined treatments were almost similar in both cell lines, although with minimal differences. Put together, these results reveal the novel synergistic effects of 2-deoxyglucose and diclofenac sodium on breast cancer cells, hence potentially elevating their pharmacological profile in the overall breast cancer therapy.
Collapse
Affiliation(s)
- Geofrey Ouma Maloba
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Tom Were
- Department of Pathology, Masinde Muliro University of Science and Technology, Kakamega 190-50100, Kenya; (T.W.); (E.B.)
| | - Erick Barasa
- Department of Pathology, Masinde Muliro University of Science and Technology, Kakamega 190-50100, Kenya; (T.W.); (E.B.)
| | - Nasreldeen Mohamed
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Arshi Arshi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, 7624 Pécs, Hungary; (G.O.M.); (N.M.); (A.A.)
| |
Collapse
|
2
|
Orobator ON, Mertens RT, Obisesan OA, Awuah SG. Energy and endoplasmic reticulum stress induction by gold(III) dithiocarbamate and 2-deoxyglucose synergistically trigger cell death in breast cancer. J Biol Chem 2024; 300:107949. [PMID: 39481597 PMCID: PMC11647619 DOI: 10.1016/j.jbc.2024.107949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
The elusiveness of triple-negative breast cancer from targeted therapy has redirected focus toward exploiting the metabolic shortcomings of these highly metastatic subtypes of breast cancer. Cueing from the metabolic heterogeneity of TNBC and the exposition of the dual dependence of some TNBCs on OXPHOS and glycolysis for ATP, we herein report the efficacy of cotreatment of TNBCs with an OXPHOS inhibitor, 2a and 2DG, a potent glycolysis inhibitor. 2a-2DG cotreatment inhibited TNBC cell proliferation with IC50 of ∼5 to 36 times lower than that of 2a alone and over 5000 times lower than IC50 of 2DG alone. 2a-2DG cotreatment suppressed mitochondrial ATP production and significantly induced AMPK activation. Mechanistic studies revealed the distinct yet synergistic contributions of 2a and 2DG to the antiproliferative effect of the cotreatment. While 2a induced apoptotic cell death, 2DG sensitized TNBCs to the antiproliferative effects of 2a via endoplasmic reticulum stress induction. Strikingly, the combination of 2a-2DG ablated SUM159 tumors in an orthotopic xenograft mouse model. This study highlights the synergistic effect of a gold-based complex with 2DG and the potential benefit of multimetabolic pathways targeting as an effective therapeutic strategy against TNBCs.
Collapse
Affiliation(s)
- Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, United States
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, United States
| | | | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, United States; Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA; Center for Bioelectronics and Nanomedicine, University of Kentucky, Lexington, Kentucky, USA.
| |
Collapse
|
3
|
Repas J, Frlic T, Snedec T, Kopitar AN, Sourij H, Janež A, Pavlin M. Physiologically Achievable Concentration of 2-Deoxy-D-Glucose Stimulates IFN-γ Secretion in Activated T Cells In Vitro. Int J Mol Sci 2024; 25:10384. [PMID: 39408714 PMCID: PMC11476708 DOI: 10.3390/ijms251910384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
2-deoxy-D-glucose (2DG) is a glycolysis and protein N-glycosylation inhibitor with promising anti-tumor and immunomodulatory effects. However, 2DG can also suppress T cell function, including IFN-γ secretion. Few human T cell studies have studied low-dose 2DG, which can increase IFN-γ in a Jurkat clone. We therefore investigated 2DG's effect on IFN-γ in activated human T cells from PBMCs, with 2DG treatment commenced either concurrently with activation or 48 h after activation. Concurrent 2DG treatment decreased IFN-γ secretion in a dose-dependent manner. However, 2DG treatment of pre-activated T cells had a hormetic effect on IFN-γ, with 0.15-0.6 mM 2DG (achievable in vivo) increasing and >2.4 mM 2DG reducing its secretion. In contrast, IL-2 levels declined monotonously with increasing 2DG concentration. Lower 2DG concentrations reduced PD-1 and increased CD69 expression regardless of treatment timing. The absence of increased T-bet or Eomes expression or IFNG transcription suggests another downstream mechanism. 2DG dose-dependently induced the unfolded protein response, suggesting a possible role in increased IFN-γ secretion, possibly by increasing the ER folding capacity for IFN-γ via increased chaperone expression. Overall, low-dose, short-term 2DG exposure could potentially improve the T cell anti-tumor response.
Collapse
Affiliation(s)
- Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tjaša Frlic
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Tadeja Snedec
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
| | - Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Medical University Graz, 8010 Graz, Austria;
| | - Andrej Janež
- Clinical Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.R.); (T.F.); (T.S.)
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Chen M, Liu Y, Li Y, Liu X. Tumor-targeted nano-assemblies for energy-blocking cocktail therapy in cancer. Acta Biomater 2024; 184:368-382. [PMID: 38908417 DOI: 10.1016/j.actbio.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/26/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
Starvation therapy aims to "starve" tumor cells by cutting off their nutritional supply. However, due to the complex and varied energy metabolism of tumors, targeting a single nutrient supply often fails to yield significant therapeutic benefits. This study proposes a tumor energy cocktail therapy that combines metformin, an oxidative phosphorylation inhibitor, with 2-deoxy-d-glucose (2-DG), a glycolysis inhibitor, to target tumor cells. To minimize the dosage of both drugs, we have developed a drug delivery strategy that prepared metformin as a nanoderivative, denoted as MA-dots. These MA-dots not only preserve the antitumor properties of metformin but also serve as a targeted delivery platform for 2-DG, ensuring its direct reach to the tumor site. Upon reaching the acidic tumor environment, the composite disintegrates, releasing 2-DG to inhibit glycolysis by targeting hexokinase 2 (HK2), the key enzyme in glycolysis, while MA-dots inhibit mitochondrial OXPHOS. This dual action significantly reduces ATP production in tumor cells, leading to apoptosis. In human lung tumor cells, the half-maximal inhibitory concentration (IC50) of 2-DG@MA-dots was significantly lower than that of either metformin or 2-DG alone, showing a nearly 100-fold and 30-fold reduction in IC50 values to 11.78 µg mL-1, from 1159 µg mL-1 and 351.20 µg mL-1, respectively. In studies with A549 tumor-bearing mice, the combination of low-dose 2-DG and metformin did not impede tumor growth, whereas 2-DG@MA-dots markedly decreased tumor volume, with the mean final tumor volume in the combination treatment group being approximately 89 times greater than that in the 2-DG@MA-dot group. STATEMENT OF SIGNIFICANCE: Metformin is a promising antitumor agent capable of modulating mitochondrial oxidative phosphorylation to inhibit cancer growth. However, its antitumor efficacy is limited when used alone due to compensatory energy mechanisms. Hence, we introduced glycolysis inhibitor 2-deoxy-d-glucose (2-DG) to inhibit an alternative tumor energy pathway. In our study, we developed a drug delivery strategy using metformin-derived nanomedicine (MA-dots) to load 2-DG. This approach enables the co-delivery of both drugs and their synergistic effect at the tumor site, disrupting both energy pathways and introducing an innovative "energy cocktail therapy".
Collapse
Affiliation(s)
- Manling Chen
- Institute of Clean Energy Chemistry, Key Laboratory for Green Synthesis and Preparative Chemistry of Advanced Materials, College of Chemistry, Liaoning University, Shenyang 110036, Liaoning, PR China
| | - Yidu Liu
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, Liaoning, PR China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, Liaoning, PR China.
| | - Xue Liu
- Institute of Clean Energy Chemistry, Key Laboratory for Green Synthesis and Preparative Chemistry of Advanced Materials, College of Chemistry, Liaoning University, Shenyang 110036, Liaoning, PR China; School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, PR Singapore.
| |
Collapse
|
5
|
Muller C, Lacroix-Malgras V, Kluza J, Laine W, Güler Y, Bost F, Boisbrun M, Mazerbourg S, Flament S. The troglitazone derivative EP13 disrupts energy metabolism through respiratory chain complex I inhibition in breast cancer cells and potentiates the antiproliferative effect of glycolysis inhibitors. Cancer Cell Int 2024; 24:132. [PMID: 38594745 PMCID: PMC11005237 DOI: 10.1186/s12935-024-03319-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The metabolism of cancer cells generally differs from that of normal cells. Indeed, most cancer cells have a high rate of glycolysis, even at normal oxygen concentrations. These metabolic properties can potentially be exploited for therapeutic intervention. In this context, we have developed troglitazone derivatives to treat hormone-sensitive and triple-negative breast cancers, which currently lack therapeutic targets, have an aggressive phenotype, and often have a worse prognosis than other subtypes. Here, we studied the metabolic impact of the EP13 compound, a desulfured derivative of Δ2-troglitazone that we synthetized and is more potent than its parent compounds. METHODS EP13 was tested on two triple-negative breast cancer cell lines, MDA-MB-231 and Hs578T, and on the luminal cell line MCF-7. The oxygen consumption rate (OCR) of the treated cell lines, Hs578T mammospheres and isolated mitochondria was measured using the XFe24 Seahorse analyser. ROS production was quantified using the MitoSOX fluorescent probe. Glycolytic activity was evaluated through measurement of the extracellular acidification rate (ECAR), glucose consumption and lactate production in extracellular medium. The synergistic effect of EP13 with glycolysis inhibitors (oxamate and 2-deoxyglucose) on cell cytotoxicity was established using the Chou-Talalay method. RESULTS After exposure to EP13, we observed a decrease in the mitochondrial oxygen consumption rate in MCF7, MDA-MB-231 and Hs578T cells. EP13 also modified the maximal OCR of Hs578T spheroids. EP13 reduced the OCR through inhibition of respiratory chain complex I. After 24 h, ATP levels in EP13-treated cells were not altered compared with those in untreated cells, suggesting compensation by glycolysis activity, as shown by the increase in ECAR, the glucose consumption and lactate production. Finally, we performed co-treatments with EP13 and glycolysis inhibitors (oxamate and 2-DG) and observed that EP13 potentiated their cytotoxic effects. CONCLUSION This study demonstrates that EP13 inhibits OXPHOS in breast cancer cells and potentiates the effect of glycolysis inhibitors.
Collapse
Affiliation(s)
- Claire Muller
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | | | - Jérôme Kluza
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pour la Recherche Sur le Cancer de Lille, UMR 9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - William Laine
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pour la Recherche Sur le Cancer de Lille, UMR 9020 - UMR-S 1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Yonca Güler
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France
| | - Frédéric Bost
- Inserm U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire, Team Cancer Metabolism, Environment, F-06200, Nice, France
| | | | - Sabine Mazerbourg
- Université de Lorraine, CNRS, CRAN, F-54000, Nancy, France.
- CRAN, UMR 7039, Faculté des Sciences et Technologies, BP 70239, 54506, Vandœuvre-lès-Nancy, France.
| | | |
Collapse
|
6
|
Slighoua M, Mahdi I, Moussaid FZ, Kamaly OA, Amrati FEZ, Conte R, Drioiche A, Saleh A, Housseini AI, Bari A, Bousta D. LC-MS/MS and GC/MS Profiling of Petroselinum sativum Hoffm. and Its Topical Application on Burn Wound Healing and Related Analgesic Potential in Rats. Metabolites 2023; 13:metabo13020260. [PMID: 36837879 PMCID: PMC9963972 DOI: 10.3390/metabo13020260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Parsley (Petroselinum sativum Hoffm.) is renowned for its ethnomedicinal uses including managing pain, wound, and dermal diseases. We previously highlighted the estrogenic and anti-inflammatory properties of parsley and profiled the phytochemistry of its polyphenolic fraction using HPLC-DAD. To extend our investigation, we here characterized the phytochemical composition of the hydro-ethanolic extract using LC-MS/MS and GC-MS upon silylation, and evaluated the antioxidant, analgesic, antimicrobial, and wound healing activities of its hydro-ethanolic and polyphenolic fraction. The antioxidant property was assessed using FRAP, DPPH, and TAC assays. The antimicrobial activity was tested against four wound infectious microbes (Staphylococcus aureus, Pseudomonas aeruginosa and Candida albicans). The analgesic effect was studied using acetic acid (counting the number of writhes) and formalin (recording the licking and biting times) injections while the wound healing activity was evaluated using burn model in vivo. The LC-MS/MS showed that the hydro-ethanolic contains four polyphenols (oleuropein, arbutin, myricetin, and naringin) while GC-MS revealed that it contains 20 compounds including malic acid, D-glucose, and galactofuranoside. The hydro-ethanolic (1000 mg/kg) decreased abdominal writhes (38.96%) and licking time (37.34%). It also elicited a strong antioxidant activity using DPPH method (IC50 = 19.38 ± 0.15 µg/mL). Polyphenols exhibited a good antimicrobial effect (MIC = 3.125-12.5 mg/mL). Moreover, both extracts showed high wound contraction by 97.17% and 94.98%, respectively. This study provides evidence that P. sativum could serve as a source of bio-compounds exhibiting analgesic effect and their promising application in mitigating ROS-related disorders, impeding wound infections, and enhancing burn healing.
Collapse
Affiliation(s)
- Meryem Slighoua
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
- Correspondence: (M.S.); (O.A.K.)
| | - Ismail Mahdi
- AgroBioSciences Research Program, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, 11, Ben-Guerir 43150, Morocco
| | - Fatima Zahrae Moussaid
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
| | - Omkulthom Al Kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
- Correspondence: (M.S.); (O.A.K.)
| | - Fatima Ez-zahra Amrati
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)—CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Aziz Drioiche
- Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay 19 Ismail University, Meknes 50000, Morocco
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Abdelilah Iraqi Housseini
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
| | - Amina Bari
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
| | - Dalila Bousta
- Laboratory of Biotechnology, Environment, Agro-Food, and Health (LBEAS), Faculty of Sciences, University Sidi-Mohamed-Ben-Abdellah (USMBA), Fez 30050, Morocco
| |
Collapse
|
7
|
Slighoua M, Chebaibi M, Mahdi I, Amrati FEZ, Conte R, Cordero MAW, Alotaibi A, Saghrouchni H, Agour A, Zair T, Bari A, Bousta D. The LC-MS/MS Identification and Analgesic and Wound Healing Activities of Lavandula officinalis Chaix: In Vivo and In Silico Approaches. PLANTS (BASEL, SWITZERLAND) 2022; 11:3222. [PMID: 36501262 PMCID: PMC9738568 DOI: 10.3390/plants11233222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
We earlier emphasized in vivo the lavender plant's (Lavandula officinalis Chaix.) anti-inflammatory and estrogenic activities and described the chemical compositions of its hydro-ethanolic (HE) extract. We used LC-MS/MS and GC-MS analyses to profile the phytochemical composition of the HE extract and to assess the analgesic and wound-healing effects of both the hydro-ethanolic (HE) and polyphenolic (LOP) extracts in vivo and in silico. The analgesic activity was studied using two methods: acetic acid and formalin injections in mice. The wound-healing activity was carried out over 25 days using a burn model in rats. In the in silico study, the polyphenols identified in the plant were docked in the active sites of three enzymes: casein kinase-1, cyclooxygenase-2, and glycogen synthase kinase-3β. The LC-MS/MS identified some phenolic compounds, mainly apigenin, catechin, and myricetin, and the GC-MS analysis revealed the presence of 19 volatile compounds with triazole, D-glucose, hydroxyphenyl, and D-Ribofuranose as the major compounds. The HE and LOP extracts showed significant decreases in abdominal writhes, and the higher licking time of the paw (57.67%) was observed using the LOP extract at 200 mg/kg. Moreover, both extracts showed high healing percentages, i.e., 99.31 and 92.88%, compared to the control groups, respectively. The molecular docking showed that myricetin, amentoflavone, apigenin, and catechin are the most active molecules against the three enzyme receptors. This study sheds light on the potential of L. officinalis Chaix as a source of natural products for pharmaceutical applications for analgesic purposes as well as their utility in promoting burn-healing activity.
Collapse
Affiliation(s)
- Meryem Slighoua
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Mohamed Chebaibi
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy of the Fez, Sidi Mohamed Ben Abdellah University, B.P. 1893, Km 22, Road of Sidi Harazem, Fez 30000, Morocco
| | - Ismail Mahdi
- AgroBioSciences Research Program, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, Ben-Guerir 43150, Morocco
| | - Fatima Ez-zahra Amrati
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)—CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Mary Anne W. Cordero
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Amal Alotaibi
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hamza Saghrouchni
- Department of Biotechnology, Institute of Natural and Applied Sciences, Çukurova University, Balcali/Sariçam, Adana 01330, Turkey
| | - Abdelkrim Agour
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and Quality of Life, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, P.O. Box 1796, Fez 30000, Morocco
| | - Touria Zair
- Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay 19 Ismail University, B.P. 11201, Meknes 50070, Morocco
| | - Amina Bari
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Dalila Bousta
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| |
Collapse
|
8
|
Mitochondrial targeting theranostic nanomedicine and molecular biomarkers for efficient cancer diagnosis and therapy. Biomed Pharmacother 2022; 153:113451. [DOI: 10.1016/j.biopha.2022.113451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
|
9
|
Khajah MA, Khushaish S, Luqmani YA. Glucose deprivation reduces proliferation and motility, and enhances the anti-proliferative effects of paclitaxel and doxorubicin in breast cell lines in vitro. PLoS One 2022; 17:e0272449. [PMID: 35917304 PMCID: PMC9345370 DOI: 10.1371/journal.pone.0272449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Background Breast cancer chemotherapy with high dose alkylating agents is severely limited by their collateral toxicity to crucial normal tissues such as immune and gut cells. Taking advantage of the selective dependence of cancer cells on high glucose and combining glucose deprivation with these agents could produce therapeutic synergy. Methods In this study we examined the effect of glucose as well as its deprivation, and antagonism using the non-metabolized analogue 2-deoxy glucose, on the proliferation of several breast cancer cell lines MCF7, MDA-MB-231, YS1.2 and pII and one normal breast cell line, using the MTT assay. Motility was quantitatively assessed using the wound healing assay. Lactate, as the end product of anaerobic glucose metabolism, secreted into culture medium was measured by a biochemical assay. The effect of paclitaxel and doxorubicin on cell proliferation was tested in the absence and presence of low concentrations of glucose using MTT assay. Results In all cell lines, glucose supplementation enhanced while glucose deprivation reduced both their proliferation and motility. Lactate added to the medium could substitute for glucose. The inhibitory effects of paclitaxel and doxorubicin were significantly enhanced when glucose concentration was decreased in the culture medium, requiring 1000-fold lesser concentration to achieve a similar degree of inhibition to that seen in glucose-containing medium. Conclusion Our data show that a synergy was obtained by combining paclitaxel and doxorubicin with glucose reduction to inhibit cancer cell growth, which in vivo, might be achieved by applying a carbohydrate-restricted diet during the limited phase of application of chemotherapy; this could permit a dose reduction of the cytotoxic agents, resulting in greater tolerance and lesser side effects.
Collapse
|
10
|
Sun X, Fan T, Sun G, Zhou Y, Huang Y, Zhang N, Zhao L, Zhong R, Peng Y. 2-Deoxy-D-glucose increases the sensitivity of glioblastoma cells to BCNU through the regulation of glycolysis, ROS and ERS pathways: In vitro and in vivo validation. Biochem Pharmacol 2022; 199:115029. [PMID: 35381210 DOI: 10.1016/j.bcp.2022.115029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022]
Abstract
Chloroethylnitrosoureas (CENUs) exert antitumor activity via producing dG-dC interstrand crosslinks (ICLs). However, tumor resistance make it necessary to find novel strategies to improve the therapeutic effect of CENUs. 2-Deoxy-D-glucose (2-DG) is a well-known glycolytic inhibitor, which can reprogram tumor energy metabolism closely related to tumor resistance. Here, we investigated the chemosensitization effect of 2-DG on l,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) against glioblastoma cells and the underlying mechanisms. We found that 2-DG significantly increased the inhibitory effects of BCNU on tumor cells compared with BCNU alone, while 2-DG showed no obvious enhancing effect on the BCNU-induced cytotoxicity for normal HaCaT and HA1800 cells. Proliferation, migration and invasion determinations presented the same trend as survival on tumor cells. 2-DG plus BCNU increased the energy deficiency through a more effective inhibition of glycolytic pathway. Notably, the combination of 2-DG and BCNU aggravated oxidative stress in glioblastoma cells, along with a significant decrease in glutathione (GSH) levels, and an increase in intracellular reactive oxygen species (ROS). Subsequently, we demonstrated that the combination treatment led to increased apoptosis via activating mitochondria and endoplasmic reticulum stress (ERS) related apoptosis pathways. Finally, we found that the dG-dC level was significantly increased after 2-DG pretreatment compared to BCNU alone by HPLC-ESI-MS/MS analysis. Finally, in vivo, 2-DG plus BCNU significantly suppressed tumor growth with lower side effects compared with BCNU alone in tumor-bearing mice. In summary, we proposed that 2-DG may have potential to increase the sensitivity of glioblastoma cells to BCNU by regulating glycolysis, ROS and ERS pathways in clinical setting.
Collapse
Affiliation(s)
- Xiaodong Sun
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Tengjiao Fan
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China; Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing 100079, China.
| | - Guohui Sun
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Yue Zhou
- Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Xian Nong Tan Street, Beijing 100050, China.
| | - Yaxin Huang
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Na Zhang
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Lijiao Zhao
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Rugang Zhong
- Beijing Key Laboratory of Environment & Viral Oncology, College of Life Science and Chemistry, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Yongzhen Peng
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, College of Environmental and Chemical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
11
|
López-Camacho E, Trilla-Fuertes L, Gámez-Pozo A, Dapía I, López-Vacas R, Zapater-Moros A, Lumbreras-Herrera MI, Arias P, Zamora P, Vara JÁF, Espinosa E. Synergistic effect of antimetabolic and chemotherapy drugs in triple-negative breast cancer. Biomed Pharmacother 2022; 149:112844. [PMID: 35339109 DOI: 10.1016/j.biopha.2022.112844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/02/2022] Open
Abstract
The triple-negative breast cancer (TNBC) subtype comprises approximately 15% of all breast cancers and is associated with poor long-term outcomes. Classical chemotherapy remains the standard of treatment, with toxicity and resistance being major limitations. TNBC is a high metabolic group, and antimetabolic drugs are effective in inhibiting TNBC cell growth. We analyzed the combined effect of chemotherapy and antimetabolic drug combinations in MDA-MB-231, MDA-MB-468 and HCC1143 human TNBC cell lines. Cells were treated with each drug or with drug combinations at a range of concentrations to establish the half-maximal inhibitory concentrations (IC50). The dose-effects of each drug or drug combination were calculated, and the synergistic or antagonistic effects of drug combinations were defined. Chemotherapy and antimetabolic drugs exhibited growth inhibitory effects on TNBC cell lines. Antimetabolic drugs targeting the glycolysis pathway had a synergistic effect with chemotherapy drugs, and antiglycolysis drug combinations also had a synergistic effect. The use of these drug combinations could lead to new therapeutic strategies that reduce chemotherapy drug doses, decreasing their toxic effect, or that maintain the doses but enhance their efficacy by their synergistic effect with other drugs.
Collapse
Affiliation(s)
- Elena López-Camacho
- Molecular Oncology Lab, La Paz University Hospital-IdiPAZ, Madrid, Spain; Biomedica Molecular Medicine SL, Madrid, Spain
| | | | - Angelo Gámez-Pozo
- Molecular Oncology Lab, La Paz University Hospital-IdiPAZ, Madrid, Spain; Biomedica Molecular Medicine SL, Madrid, Spain
| | - Irene Dapía
- Pharmacogenetics Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Autonomous University of Madrid, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases-CIBERER, ISCIII, Madrid, Spain
| | - Rocío López-Vacas
- Molecular Oncology Lab, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Andrea Zapater-Moros
- Biomedica Molecular Medicine SL, Madrid, Spain; Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | | | - Pedro Arias
- Pharmacogenetics Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Autonomous University of Madrid, Madrid, Spain; Biomedical Research Networking Center on Rare Diseases-CIBERER, ISCIII, Madrid, Spain
| | - Pilar Zamora
- Medical Oncology Service, La Paz University Hospital, Madrid, Spain
| | - Juan Ángel Fresno Vara
- Molecular Oncology Lab, La Paz University Hospital-IdiPAZ, Madrid, Spain; Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Enrique Espinosa
- Medical Oncology Service, La Paz University Hospital, Madrid, Spain; Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain.
| |
Collapse
|
12
|
Hellemann E, Walker JL, Lesko MA, Chandrashekarappa DG, Schmidt MC, O’Donnell AF, Durrant JD. Novel mutation in hexokinase 2 confers resistance to 2-deoxyglucose by altering protein dynamics. PLoS Comput Biol 2022; 18:e1009929. [PMID: 35235554 PMCID: PMC8920189 DOI: 10.1371/journal.pcbi.1009929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/14/2022] [Accepted: 02/16/2022] [Indexed: 01/16/2023] Open
Abstract
Glucose is central to many biological processes, serving as an energy source and a building block for biosynthesis. After glucose enters the cell, hexokinases convert it to glucose-6-phosphate (Glc-6P) for use in anaerobic fermentation, aerobic oxidative phosphorylation, and the pentose-phosphate pathway. We here describe a genetic screen in Saccharomyces cerevisiae that generated a novel spontaneous mutation in hexokinase-2, hxk2G238V, that confers resistance to the toxic glucose analog 2-deoxyglucose (2DG). Wild-type hexokinases convert 2DG to 2-deoxyglucose-6-phosphate (2DG-6P), but 2DG-6P cannot support downstream glycolysis, resulting in a cellular starvation-like response. Curiously, though the hxk2G238V mutation encodes a loss-of-function allele, the affected amino acid does not interact directly with bound glucose, 2DG, or ATP. Molecular dynamics simulations suggest that Hxk2G238V impedes sugar binding by altering the protein dynamics of the glucose-binding cleft, as well as the large-scale domain-closure motions required for catalysis. These findings shed new light on Hxk2 dynamics and highlight how allosteric changes can influence catalysis, providing new structural insights into this critical regulator of carbohydrate metabolism. Given that hexokinases are upregulated in some cancers and that 2DG and its derivatives have been studied in anti-cancer trials, the present work also provides insights that may apply to cancer biology and drug resistance. Glucose fuels many of the energy-production processes required for normal cell growth. Before glucose can participate in these processes, it must first be chemically modified by proteins called hexokinases. To better understand how hexokinases modify glucose—and how mutations in hexokinase genes might confer drug resistance—we evolved resistance in yeast to a toxic hexokinase-binding molecule called 2DG. We discovered a mutation in the hexokinase gene that confers 2DG resistance and reduces the protein’s ability to modify glucose. Biochemical analyses and computer simulations of the hexokinase protein suggest that the mutation diminishes glucose binding by altering enzyme flexibility. This work shows how cells can evolve resistance to toxins via only modest changes to protein structures. Furthermore, because cancer-cell hexokinases are particularly active, 2DG has been studied as cancer chemotherapy. Thus, the insights this work provides might also apply to cancer biology.
Collapse
Affiliation(s)
- Erich Hellemann
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jennifer L. Walker
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mitchell A. Lesko
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Dakshayini G. Chandrashekarappa
- University of Pittsburgh School of Medicine, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Martin C. Schmidt
- University of Pittsburgh School of Medicine, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Allyson F. O’Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (AFO); (JDD)
| | - Jacob D. Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (AFO); (JDD)
| |
Collapse
|
13
|
Metformin and Breast Cancer: Where Are We Now? Int J Mol Sci 2022; 23:ijms23052705. [PMID: 35269852 PMCID: PMC8910543 DOI: 10.3390/ijms23052705] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most prevalent cancer and the leading cause of cancer-related death among women worldwide. Type 2 diabetes–associated metabolic traits such as hyperglycemia, hyperinsulinemia, inflammation, oxidative stress, and obesity are well-known risk factors for breast cancer. The insulin sensitizer metformin, one of the most prescribed oral antidiabetic drugs, has been suggested to function as an antitumoral agent, based on epidemiological and retrospective clinical data as well as preclinical studies showing an antiproliferative effect in cultured breast cancer cells and animal models. These benefits provided a strong rationale to study the effects of metformin in routine clinical care of breast cancer patients. However, the initial enthusiasm was tempered after disappointing results in randomized controlled trials, particularly in the metastatic setting. Here, we revisit the current state of the art of metformin mechanisms of action, critically review past and current metformin-based clinical trials, and briefly discuss future perspectives on how to incorporate metformin into the oncologist’s armamentarium for the prevention and treatment of breast cancer.
Collapse
|
14
|
Repas J, Zügner E, Gole B, Bizjak M, Potočnik U, Magnes C, Pavlin M. Metabolic profiling of attached and detached metformin and 2-deoxy-D-glucose treated breast cancer cells reveals adaptive changes in metabolome of detached cells. Sci Rep 2021; 11:21354. [PMID: 34725457 PMCID: PMC8560930 DOI: 10.1038/s41598-021-98642-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/06/2021] [Indexed: 02/05/2023] Open
Abstract
Anchorage-independent growth of cancer cells in vitro is correlated to metastasis formation in vivo. Metformin use is associated with decreased breast cancer incidence and currently evaluated in cancer clinical trials. The combined treatment with metformin and 2-deoxy-D-glucose (2DG) in vitro induces detachment of viable MDA-MB-231 breast cancer cells that retain their proliferation capacity. This might be important for cell detachment from primary tumors, but the metabolic changes involved are unknown. We performed LC/MS metabolic profiling on separated attached and detached MDA-MB-231 cells treated with metformin and/or 2DG. High 2DG and metformin plus 2DG altered the metabolic profile similarly to metformin, inferring that metabolic changes are necessary but not sufficient while the specific effects of 2DG are crucial for detachment. Detached cells had higher NADPH levels and lower fatty acids and glutamine levels compared to attached cells, supporting the role of AMPK activation and reductive carboxylation in supporting anchorage-independent survival. Surprisingly, the metabolic profile of detached cells was closer to untreated control cells than attached treated cells, suggesting detachment might help cells adapt to energy stress. Metformin treated cells had higher fatty and amino acid levels with lower purine nucleotide levels, which is relevant for understanding the anticancer mechanisms of metformin.
Collapse
Affiliation(s)
- Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Elmar Zügner
- Joanneum Research Health - Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Boris Gole
- Center for Human Molecular Genetics and Pharmacogenomics, Medical Faculty, University of Maribor, Maribor, Slovenia
| | - Maruša Bizjak
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
- Pharmacy Institute, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Uroš Potočnik
- Center for Human Molecular Genetics and Pharmacogenomics, Medical Faculty, University of Maribor, Maribor, Slovenia
- Laboratory for Biochemistry, Molecular biology and Genomics, Faculty for Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Christoph Magnes
- Joanneum Research Health - Institute for Biomedicine and Health Sciences, Graz, Austria.
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
15
|
Inhibition of Metabolism as a Therapeutic Option for Tamoxifen-Resistant Breast Cancer Cells. Cells 2021; 10:cells10092398. [PMID: 34572047 PMCID: PMC8467413 DOI: 10.3390/cells10092398] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer cells have an increased need for glucose and, despite aerobic conditions, obtain their energy through aerobic oxidation and lactate fermentation, instead of aerobic oxidation alone. Glutamine is an essential amino acid in the human body. Glutaminolysis and glycolysis are crucial for cancer cell survival. In the therapy of estrogen receptor α (ERα)-positive breast cancer (BC), the focus lies on hormone sensitivity targeting therapy with selective estrogen receptor modulators (SERMs) such as 4-hydroxytamoxifen (4-OHT), although this therapy is partially limited by the development of resistance. Therefore, further targets for therapy improvement of ERα-positive BC with secondary 4-OHT resistance are needed. Hence, increased glucose requirement and upregulated glutaminolysis in BC cells could be used. We have established sublines of ERα-positive MCF7 and T47D BC cells, which were developed to be resistant to 4-OHT. Further, glycolysis inhibitor 2-Deoxy-D-Glucose (2-DG) and glutaminase inhibitor CB-839 were analyzed. Co-treatments using 4-OHT and CB-839, 2-DG and CB-839, or 4-OHT, 2-DG and CB-839, respectively, showed significantly stronger inhibitory effects on viability compared to single treatments. It could be shown that tamoxifen-resistant BC cell lines, compared to the non-resistant cell lines, exhibited a stronger reducing effect on cell viability under co-treatments. In addition, the tamoxifen-resistant BC cell lines showed increased expression of proto-oncogene c-Myc compared to the parental cell lines. This could be reduced depending on the treatment. Suppression of c-Myc expression using specific siRNA completely abolished resistance to 4OH-tamoxifen. In summary, our data suggest that combined treatments affecting the metabolism of BC are suitable depending on the cellularity and resistance status. In addition, the anti-metabolic treatments affected the expression of the proto-oncogene c-Myc, a key player in the regulation of cancer cell metabolism.
Collapse
|
16
|
Sun X, Peng Y, Zhao J, Xie Z, Lei X, Tang G. Discovery and development of tumor glycolysis rate-limiting enzyme inhibitors. Bioorg Chem 2021; 112:104891. [PMID: 33940446 DOI: 10.1016/j.bioorg.2021.104891] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022]
Abstract
Tumor cells mainly provide necessary energy and substances for rapid cell growth through aerobic perglycolysis rather than oxidative phosphorylation. This phenomenon is called the "Warburg effect". The mechanism of glycolysis in tumor cells is more complicated, which is caused by the comprehensive regulation of multiple factors. Abnormal enzyme metabolism is one of the main influencing factors and inhibiting the three main rate-limiting enzymes in glycolysis is thought to be important strategy for cancer treatment. Therefore, numerous inhibitors of glycolysis rate-limiting enzyme have been developed in recent years, such as the latest HKII inhibitor and PKM2 inhibitor Pachymic acid (PA) and N-(4-(3-(3-(methylamino)-3-oxopropyl)-5-(4'-(trifluoromethyl)-[1,1'-biphenyl]-4-yl)-1H-pyrazol-1-yl)phenyl)propiolamide. The review focuses on source, structure-activity relationship, bioecological activity and mechanism of the three main rate-limiting enzymes inhibitors, and hopes to guide the future research on the design and synthesis of rate-limiting enzyme inhibitors.
Collapse
Affiliation(s)
- Xueyan Sun
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Yijiao Peng
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Jingduo Zhao
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China
| | - Zhizhong Xie
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China
| | - Xiaoyong Lei
- Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang City, PR China; Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hengyang City, Hunan Province, PR China.
| |
Collapse
|
17
|
Mack N, Mazzio E, Badisa R, Soliman KFA. Metabolic Response to the Mitochondrial Toxin 1-Methyl-4-phenylpyridinium (MPP+) in LDH-A/B Double-knockout LS174T Colon Cancer Cells. Cancer Genomics Proteomics 2021; 18:385-405. [PMID: 33994363 DOI: 10.21873/cgp.20267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Rapid glycolytic substrate-level phosphorylation (SLP) and accumulation of lactic acid are characteristics of diverse cancers. Recent advances in drug discovery have included the use of glycolytic inhibitors with mitochondrial targeting drugs to attempt to invoke an energy crisis in aggressive metabolically active chemo-resistant cancers. In this work, we examine the consequences of inhibiting mitochondrial oxidative phosphorylation (OXPHOS) with 1-methyl-4-phenylpyridinium (MPP+) in LS14T colon cancer cells containing a genetic double knock out (DKO) of lactic acid dehydrogenase (LDHA and LDHB). MATERIALS AND METHODS Several metabolic parameters were evaluated concomitant to whole transcriptomic (WT) mRNA, microRNA, and long intergenic non-coding RNAs using Affymetrix 2.1 human ST arrays. RESULTS MPP+ effectively blocked OXPHOS where a compensatory shift toward anaerobic SLP was only observed in the control vector (CV), and not observed in the LDH-A/B DKOs (lacking the ability to produce lactic acid). Despite this, there was an unexpected resilience to MPP+ in the latter in terms of energy, which displayed significantly higher resting baseline respiratory OXPHOS capacity relative to controls. At the transcriptome level, MPP+ invoked 1738 differential expressed genes (DEGs) out of 48,226; LDH-A/B DKO resulted in 855 DEGs while 349 DEGs were found to be overlapping in both groups versus respective controls, including loss of mitochondrial complex I (subunits 3 and 6), cell cycle transcripts and fluctuations in epigenetic chromatin remodeling systems. In terms of energy, the effects of MPP+ in the CV transcripts reflect the funneling of carbon intermediates toward glycolysis. The LDH-A/B DKO transcripts reflect a flow of carbons away from glycolysis toward the production of acetyl-CoA. CONCLUSION The findings from this study suggest a metabolic resilience to MPP+ in cancer cells devoid of LDH-A/B, explainable in-part by higher baseline OXPHOS respiratory ATP production, necessitating more toxin to suppress the electron transport chain.
Collapse
Affiliation(s)
- Nzinga Mack
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Elizabeth Mazzio
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Ramesh Badisa
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F A Soliman
- Pharmaceutical Sciences Division, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
18
|
Arundhathi JRD, Mathur SR, Gogia A, Deo SVS, Mohapatra P, Prasad CP. Metabolic changes in triple negative breast cancer-focus on aerobic glycolysis. Mol Biol Rep 2021; 48:4733-4745. [PMID: 34047880 DOI: 10.1007/s11033-021-06414-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023]
Abstract
Among breast cancer subtypes, the triple negative breast cancer (TNBC) has the worst prognosis. In absence of any permitted targeted therapy, standard chemotherapy is the mainstay for TNBC treatment. Hence, there is a crucial need to identify potential druggable targets in TNBCs for its effective treatment. In recent times, metabolic reprogramming has emerged as cancer cells hallmark, wherein cancer cells display discrete metabolic phenotypes to fuel cell progression and metastasis. Altered glycolysis is one such phenotype, in which even in oxygen abundance majority of cancer cells harvest considerable amount of energy through elevated glycolytic-flux. In the present review, we attempt to summarize the role of key glycolytic enzymes i.e. HK, Hexokinase; PFK, Phosphofructokinase; PKM2, Pyruvate kinase isozyme type 2; and LDH, Lactate dehydrogenase in TNBCs, and possible therapeutic options presently available.
Collapse
Affiliation(s)
- J R Dev Arundhathi
- Department of Medical Oncology, Dr BRA IRCH, AIIMS, New Delhi, 110029, India
| | - Sandeep R Mathur
- Department of Pathology, Dr BRA IRCH, AIIMS, New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, Dr BRA IRCH, AIIMS, New Delhi, 110029, India
| | - S V S Deo
- Department of Surgical Oncology, Dr BRA IRCH, AIIMS, New Delhi, 110029, India
| | | | | |
Collapse
|
19
|
Chen H, Li K, Li Y, Xie P, He J, Zhang H. An integrative pan-cancer analysis of COPB1 based on data mining. Cancer Biomark 2021; 30:13-27. [PMID: 32986658 DOI: 10.3233/cbm-200398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer will become the leading cause of death worldwide in the 21st century, meanwhile, immunotherapy is the most popular cancer treatment method in recent years. COPI Coat Complex Subunit Beta 1 (COPB1) relates to human innate immunity. However, the role of COPB1 in pan-cancer remains unclear. OBJECTIVE The purpose of this study was to explore the relationship between COPB1 mRNA expression and tumor infiltrating lymphocytes and immune examination sites in pan-cancer. METHODS Data from multiple online databases were collected. The BioGPS, UALCAN Database, COSMIC, cBioPortal, Cancer Regulome tools, Kaplan-Meier Plotter and TIMER website were utilized to perform the analysis. RESULTS Upregulation of COPB1 has been widely observed in tumor tissues compared with normal tissues. Although COPB1 has poor prognosis in pan-cancer, COPB1 high expression was beneficial to the survival of ESCA patients. Unlike ESCA, COPB1 expression in STAD was positively correlated with tumor infiltrating lymphocytes, including B cells, CD8+ T cells, neutrophils, macrophages, and dendritic cells. Finally, we also found that the expression of COPB1 in STAD was positively correlated with PD-L1 and CTLA4. CONCLUSIONS COPB1 may be a prognostic biomarker for pan-carcinoma, and also provide an immune anti-tumor strategy for STAD based on the expression of COPB1.
Collapse
|
20
|
Wanyan Y, Xu X, Liu K, Zhang H, Zhen J, Zhang R, Wen J, Liu P, Chen Y. 2-Deoxy-d-glucose Promotes Buforin IIb-Induced Cytotoxicity in Prostate Cancer DU145 Cells and Xenograft Tumors. Molecules 2020; 25:E5778. [PMID: 33297583 PMCID: PMC7730206 DOI: 10.3390/molecules25235778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 01/15/2023] Open
Abstract
Inhibition of the glycolytic pathway is a critical strategy in anticancer therapy because of the role of aerobic glycolysis in cancer cells. The glycolytic inhibitor 2-Deoxy-d-glucose (2-DG) has shown potential in combination with other anticancer agents. Buforin IIb is an effective antimicrobial peptide (AMP) with broad-spectrum anticancer activity and selectivity. The efficacy of combination treatment with 2-DG and buforin IIb in prostate cancer remains unknown. Here, we tested the efficacy of buforin IIb as a mitochondria-targeting AMP in the androgen-independent human prostate cancer cell line DU145. Combining 2-DG with buforin IIb had a synergistic toxic effect on DU145 cells and mouse xenograft tumors. Combination treatment with 2-DG and buforin IIb caused stronger proliferation inhibition, greater G1 cell cycle arrest, and higher apoptosis than either treatment alone. Combination treatment dramatically decreased L-lactate production and intracellular ATP levels, indicating severe inhibition of glycolysis and ATP production. Flow cytometry and confocal laser scanning microscopy results indicate that 2-DG may increase buforin IIb uptake by DU145 cells, thereby increasing the mitochondria-targeting capacity of buforin IIb. This may partly explain the effect of combination treatment on enhancing buforin IIb-induced apoptosis. Consistently, 2-DG increased mitochondrial dysfunction and upregulated Bax/Bcl-2, promoting cytochrome c release to initiate procaspase 3 cleavage induced by buforin IIb. These results suggest that 2-DG sensitizes prostate cancer DU145 cells to buforin IIb. Moreover, combination treatment caused minimal hemolysis and cytotoxicity to normal WPMY-1 cells. Collectively, the current study demonstrates that dual targeting of glycolysis and mitochondria by 2-DG and buforin IIb may be an effective anticancer strategy for the treatment of some advanced prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuqing Chen
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, Life Sciences College, Nanjing Normal University, Nanjing 210000, China; (Y.W.); (X.X.); (K.L.); (H.Z.); (J.Z.); (R.Z.); (J.W.); (P.L.)
| |
Collapse
|
21
|
Tilekar K, Upadhyay N, Iancu CV, Pokrovsky V, Choe JY, Ramaa CS. Power of two: combination of therapeutic approaches involving glucose transporter (GLUT) inhibitors to combat cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188457. [PMID: 33096154 PMCID: PMC7704680 DOI: 10.1016/j.bbcan.2020.188457] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
Cancer research of the Warburg effect, a hallmark metabolic alteration in tumors, focused attention on glucose metabolism whose targeting uncovered several agents with promising anticancer effects at the preclinical level. These agents' monotherapy points to their potential as adjuvant combination therapy to existing standard chemotherapy in human trials. Accordingly, several studies on combining glucose transporter (GLUT) inhibitors with chemotherapeutic agents, such as doxorubicin, paclitaxel, and cytarabine, showed synergistic or additive anticancer effects, reduced chemo-, radio-, and immuno-resistance, and reduced toxicity due to lowering the therapeutic doses required for desired chemotherapeutic effects, as compared with monotherapy. The combinations have been specifically effective in treating cancer glycolytic phenotypes, such as pancreatic and breast cancers. Even combining GLUT inhibitors with other glycolytic inhibitors and energy restriction mimetics seems worthwhile. Though combination clinical trials are in the early phase, initial results are intriguing. The various types of GLUTs, their role in cancer progression, GLUT inhibitors, and their anticancer mechanism of action have been reviewed several times. However, utilizing GLUT inhibitors as combination therapeutics has received little attention. We consider GLUT inhibitors agents that directly affect glucose transporters by binding to them or indirectly alter glucose transport by changing the transporters' expression level. This review mainly focuses on summarizing the effects of various combinations of GLUT inhibitors with other anticancer agents and providing a perspective on the current status.
Collapse
Affiliation(s)
- Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Cristina V. Iancu
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Vadim Pokrovsky
- Laboratory of Combined Therapy, N.N. Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People’s Friendship University, Moscow, Russia
| | - Jun-yong Choe
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - C. S. Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| |
Collapse
|
22
|
Shuvalov O, Fedorova O, Tananykina E, Gnennaya Y, Daks A, Petukhov A, Barlev NA. An Arthropod Hormone, Ecdysterone, Inhibits the Growth of Breast Cancer Cells via Different Mechanisms. Front Pharmacol 2020; 11:561537. [PMID: 33192507 PMCID: PMC7663021 DOI: 10.3389/fphar.2020.561537] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/30/2020] [Indexed: 12/30/2022] Open
Abstract
Ecdysterone (Ecdy) is a hormone found in arthropods, which regulates their development. It is also synthesized by a number of plants to combat insect pests. It provides a number of beneficial pharmacological effects including the anabolic and adaptogenic ones. Ecdysterone is widely marketed as food supplement to enhance the physical performance of athletes. In addition to the estrogen receptor beta (ERbeta)-dependent anabolic effect of Ecdy in muscles, the molecular mechanisms of the plethora of other Ecdy-induced pharmacological effects remain unknown. The aim of this study was to investigate the pharmacological effect of ecdysterone on human breast cancer cell lines of different molecular subtypes. Surprisingly, in contrast to the anabolic effect on muscle tissues, we have revealed a tumor suppressive effect of Ecdy on a panel of breast cancer cell lines studied. Using the SeaHorse-based energy profiling, we have demonstrated that Ecdy dampened glycolysis and respiration, as well as greatly reduced the metabolic potential of triple negative breast cancer cell lines. Furthermore, we have revealed that Ecdy strongly induced autophagy. As part of the combined treatment, based on the Combination Index (CI) and Dose Reduction Index (DRI), Ecdy synergized with doxorubicin to induce cell death in several breast cancer cell lines. In contrast, Ecdy had only minor effect on non-transformed human fibroblasts. Collectively, our results indicate that ecdysterone can be considered as a new potential adjuvant for genotoxic therapy in treatment of breast cancer patients.
Collapse
Affiliation(s)
- O Shuvalov
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia
| | - O Fedorova
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia
| | - E Tananykina
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia
| | - Y Gnennaya
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia
| | - A Daks
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia
| | - A Petukhov
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia.,Almazov National Medical Research Centre, St-Petersburg, Russia
| | - N A Barlev
- Institute of cytology, Russian Academy of Sciences (RAS), St-Petersburg, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,Orekhovich Institute of Biochemical Medicine, Moscow, Russia
| |
Collapse
|
23
|
Fujita M, Imadome K, Somasundaram V, Kawanishi M, Karasawa K, Wink DA. Metabolic characterization of aggressive breast cancer cells exhibiting invasive phenotype: impact of non-cytotoxic doses of 2-DG on diminishing invasiveness. BMC Cancer 2020; 20:929. [PMID: 32993545 PMCID: PMC7525976 DOI: 10.1186/s12885-020-07414-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/15/2020] [Indexed: 01/08/2023] Open
Abstract
Background Metabolic reprogramming is being recognized as a fundamental hallmark of cancer, and efforts to identify drugs that can target cancer metabolism are underway. In this study, we used human breast cancer (BC) cell lines and established their invading phenotype (INV) collected from transwell inserts to compare metabolome differences and evaluate prognostic significance of the metabolome in aggressive BC invasiveness. Methods The invasiveness of seven human BC cell lines were compared using the transwell invasion assay. Among these, INV was collected from SUM149, which exhibited the highest invasiveness. Levels of metabolites in INV were compared with those of whole cultured SUM149 cells (WCC) using CE-TOFMS. The impact of glycolysis in INV was determined by glucose uptake assay using fluorescent derivative of glucose (2-NBDG), and significance of glycolysis, or tricarboxylic acid cycle (TCA) and electron transport chain (ETC) in the invasive process were further determined in aggressive BC cell lines, SUM149, MDA-MB-231, HCC1937, using invasion assays in the presence or absence of inhibitors of glycolysis, TCA cycle or ETC. Results SUM149 INV sub-population exhibited a persistent hyperinvasive phenotype. INV were hyper-glycolytic with increased glucose (2-NBDG) uptake; diminished glucose-6-phosphate (G6P) levels but elevated pyruvate and lactate, along with higher expression of phosphorylated-pyruvate dehydrogenase (pPDH) compared to WCC. Notably, inhibiting of glycolysis with lower doses of 2-DG (1 mM), non-cytotoxic to MDA-MB-231 and HCC1937, was effective in diminishing invasiveness of aggressive BC cell lines. In contrast, 3-Nitropropionic acid (3-NA), an inhibitor of succinate dehydrogenase, the enzyme that oxidizes succinate to fumarate in TCA cycle, and functions as complex II of ETC, had no significant effect on their invasiveness, although levels of TCA metabolites or detection of mitochondrial membrane potential with JC-1 staining, indicated that INV cells originally had functional TCA cycles and membrane potential. Conclusions Hyper-glycolytic phenotype of invading cells caters to rapid energy production required for invasion while TCA cycle/ETC cater to cellular energy needs for sustenance in aggressive BC. Lower, non-cytotoxic doses of 2-DG can hamper invasion and can potentially be used as an adjuvant with other anti-cancer therapies without the usual side-effects associated with cytotoxic doses.
Collapse
Affiliation(s)
- Mayumi Fujita
- Department of Basic Medical Science for Radiation Damages, National Institute of Radiological Sciences, NIRS, National Institute for Quantum and Radiological Science and Technology, QST, 4-9-1, Anagawa, Inage-ku, Chiba-shi, Chiba-ken, Japan.
| | - Kaori Imadome
- Department of Basic Medical Science for Radiation Damages, National Institute of Radiological Sciences, NIRS, National Institute for Quantum and Radiological Science and Technology, QST, 4-9-1, Anagawa, Inage-ku, Chiba-shi, Chiba-ken, Japan
| | - Veena Somasundaram
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Miki Kawanishi
- Department of Radiation Oncology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kumiko Karasawa
- Department of Radiation Oncology, Tokyo Women's Medical University, Tokyo, Japan
| | - David A Wink
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
24
|
Lü J, Zhang C, Han J, Xu Z, Li Y, Zhen L, Zhao Q, Guo Y, Wang Z, Bischof E, Yu Z. Starvation stress attenuates the miRNA-target interaction in suppressing breast cancer cell proliferation. BMC Cancer 2020; 20:627. [PMID: 32631271 PMCID: PMC7339532 DOI: 10.1186/s12885-020-07118-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/26/2020] [Indexed: 11/10/2022] Open
Abstract
Background Emerging evidence has demonstrated the limited access to metabolic substrates as an effective approach to block cancer cell growth. The mechanisms remain unclear. Our previous work has revealed that miR-221/222 plays important role in regulating breast cancer development and progression through interaction with target gene p27. Results Herein, we determined the miRNA-mRNA interaction in breast cancer cells under induced stress status of starvation. Starvation stimulation attenuated the miR-221/222-p27 interaction in MDA-MB-231 cells, thereby increased p27 expression and suppressed cell proliferation. Through overexpression or knockdown of miR-221/222, we found that starvation-induced stress attenuated the negative regulation of p27 expression by miR-221/222. Similar patterns for miRNA-target mRNA interaction were observed between miR-17-5p and CyclinD1, and between mR-155 and Socs1. Expression of Ago2, one of the key components of RNA-induced silencing complex (RISC), was decreased under starvation-induced stress status, which took responsibility for the impaired miRNA-target interaction since addition of exogenous Ago2 into MDA-MB-231 cells restored the miR-221/222-p27 interaction in starvation condition. Conclusions We demonstrated the attenuated interaction between miR-221/222 and p27 by starvation-induced stress in MDA-MB-231 breast cancer cells. The findings add a new page to the general knowledge of negative regulation of gene expression by miRNAs, also demonstrate a novel mechanism through which limited access to nutrients suppresses cancer cell proliferation. These insights provide a basis for development of novel therapeutic options for breast cancer.
Collapse
Affiliation(s)
- Jinhui Lü
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Chuyi Zhang
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Junyi Han
- Department of Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhen Xu
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuan Li
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lixiao Zhen
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Qian Zhao
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuefan Guo
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhaohui Wang
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Jinzhou Medical University, Liaoning, China
| | - Evelyne Bischof
- Shanghai University of Medicine and Health Sciences Clinical Medicine Division, Shanghai, China. .,Division of Internal Medicine, University Hospital of Basel, Petersgraben 4, 4051, Basel l, Switzerland.
| | - Zuoren Yu
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
25
|
Competitive glucose metabolism as a target to boost bladder cancer immunotherapy. Nat Rev Urol 2020; 17:77-106. [PMID: 31953517 DOI: 10.1038/s41585-019-0263-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2019] [Indexed: 12/24/2022]
Abstract
Bladder cancer - the tenth most frequent cancer worldwide - has a heterogeneous natural history and clinical behaviour. The predominant histological subtype, urothelial bladder carcinoma, is characterized by high recurrence rates, progression and both primary and acquired resistance to platinum-based therapy, which impose a considerable economic burden on health-care systems and have substantial effects on the quality of life and the overall outcomes of patients with bladder cancer. The incidence of urothelial tumours is increasing owing to population growth and ageing, so novel therapeutic options are vital. Based on work by The Cancer Genome Atlas project, which has identified targetable vulnerabilities in bladder cancer, immune checkpoint inhibitors (ICIs) have arisen as an effective alternative for managing advanced disease. However, although ICIs have shown durable responses in a subset of patients with bladder cancer, the overall response rate is only ~15-25%, which increases the demand for biomarkers of response and therapeutic strategies that can overcome resistance to ICIs. In ICI non-responders, cancer cells use effective mechanisms to evade immune cell antitumour activity; the overlapping Warburg effect machinery of cancer and immune cells is a putative determinant of the immunosuppressive phenotype in bladder cancer. This energetic interplay between tumour and immune cells leads to metabolic competition in the tumour ecosystem, limiting nutrient availability and leading to microenvironmental acidosis, which hinders immune cell function. Thus, molecular hallmarks of cancer cell metabolism are potential therapeutic targets, not only to eliminate malignant cells but also to boost the efficacy of immunotherapy. In this sense, integrating the targeting of tumour metabolism into immunotherapy design seems a rational approach to improve the therapeutic efficacy of ICIs.
Collapse
|
26
|
Martin SD, McGee SL. A systematic flux analysis approach to identify metabolic vulnerabilities in human breast cancer cell lines. Cancer Metab 2019; 7:12. [PMID: 31890204 PMCID: PMC6935091 DOI: 10.1186/s40170-019-0207-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023] Open
Abstract
Background Increased flux through both glycolytic and oxidative metabolic pathways is a hallmark of breast cancer cells and is critical for their growth and survival. As such, targeting this metabolic reprograming has received much attention as a potential treatment approach. However, the heterogeneity of breast cancer cell metabolism, even within classifications, suggests a necessity for an individualised approach to treatment in breast cancer patients. Methods The metabolic phenotypes of a diverse panel of human breast cancer cell lines representing the major breast cancer classifications were assessed using real-time metabolic flux analysis. Flux linked to ATP production, pathway reserve capacities and specific macromolecule oxidation rates were quantified. Suspected metabolic vulnerabilities were targeted with specific pathway inhibitors, and relative cell viability was assessed using the crystal violet assay. Measures of AMPK and mTORC1 activity were analysed through immunoblotting. Results Breast cancer cells displayed heterogeneous energy requirements and utilisation of non-oxidative and oxidative energy-producing pathways. Quantification of basal glycolytic and oxidative reserve capacities identified cell lines that were highly dependent on individual pathways, while assessment of substrate oxidation relative to total oxidative capacity revealed cell lines that were highly dependent on individual macromolecules. Based on these findings, mild mitochondrial inhibition in ESH-172 cells, including with the anti-diabetic drug metformin, and mild glycolytic inhibition in Hs578T cells reduced relative viability, which did not occur in non-transformed MCF10a cells. The effects on viability were associated with AMPK activation and inhibition of mTORC1 signalling. Hs578T were also found to be highly dependent on glutamine oxidation and inhibition of this process also impacted viability. Conclusions Together, these data highlight that systematic flux analysis in breast cancer cells can identify targetable metabolic vulnerabilities, despite heterogeneity in metabolic profiles between individual cancer cell lines.
Collapse
Affiliation(s)
- Sheree D Martin
- Metabolic Reprogramming Laboratory, Metabolic Research Unit, School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria Australia
| | - Sean L McGee
- Metabolic Reprogramming Laboratory, Metabolic Research Unit, School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria Australia
| |
Collapse
|
27
|
Samuel SM, Varghese E, Kubatka P, Triggle CR, Büsselberg D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules 2019; 9:E846. [PMID: 31835318 PMCID: PMC6995629 DOI: 10.3390/biom9120846] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/25/2022] Open
Abstract
Interest has grown in studying the possible use of well-known anti-diabetic drugs as anti-cancer agents individually or in combination with, frequently used, chemotherapeutic agents and/or radiation, owing to the fact that diabetes heightens the risk, incidence, and rapid progression of cancers, including breast cancer, in an individual. In this regard, metformin (1, 1-dimethylbiguanide), well known as 'Glucophage' among diabetics, was reported to be cancer preventive while also being a potent anti-proliferative and anti-cancer agent. While meta-analysis studies reported a lower risk and incidence of breast cancer among diabetic individuals on a metformin treatment regimen, several in vitro, pre-clinical, and clinical studies reported the efficacy of using metformin individually as an anti-cancer/anti-tumor agent or in combination with chemotherapeutic drugs or radiation in the treatment of different forms of breast cancer. However, unanswered questions remain with regards to areas such as cancer treatment specific therapeutic dosing of metformin, specificity to cancer cells at high concentrations, resistance to metformin therapy, efficacy of combinatory therapeutic approaches, post-therapeutic relapse of the disease, and efficacy in cancer prevention in non-diabetic individuals. In the current article, we discuss the biology of metformin and its molecular mechanism of action, the existing cellular, pre-clinical, and clinical studies that have tested the anti-tumor potential of metformin as a potential anti-cancer/anti-tumor agent in breast cancer therapy, and outline the future prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer drug in the treatment of breast cancer.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
28
|
Muniraj N, Siddharth S, Nagalingam A, Walker A, Woo J, Győrffy B, Gabrielson E, Saxena NK, Sharma D. Withaferin A inhibits lysosomal activity to block autophagic flux and induces apoptosis via energetic impairment in breast cancer cells. Carcinogenesis 2019; 40:1110-1120. [PMID: 30698683 PMCID: PMC10893887 DOI: 10.1093/carcin/bgz015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/02/2019] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
Withaferin A (WFA), a steroidal lactone, negatively regulates breast cancer growth however, its mechanisms of action remain largely elusive. We found that WFA blocks autophagy flux and lysosomal proteolytic activity in breast cancer cells. WFA increases accumulation of autophagosomes, LC3B-II conversion, expression of autophagy-related proteins and autophagosome/lysosome fusion. Autolysosomes display the characteristics of acidic compartments in WFA-treated cells; however, the protein degradation activity of lysosomes is inhibited. Blockade of autophagic flux reduces the recycling of cellular fuels leading to insufficient substrates for tricarboxylic acid (TCA) cycle and impaired oxidative phosphorylation. WFA decreases expression and phosphorylation of lactate dehydrogenase, the key enzyme that catalyzes pyruvate-to-lactate conversion, reduces adenosine triphosphate levels and increases AMP-activated protein kinase (AMPK) activation. AMPK inhibition abrogates while AMPK activation potentiates WFA's effect. WFA and 2-deoxy-d-glucose combination elicits synergistic inhibition of breast cancer cells. Genetic knockout of BECN1 and ATG7 fails to rescue cells from WFA treatment; in contrast, addition of methyl pyruvate to supplement TCA cycle protects WFA-treated cells. Together, these results implicate that WFA is a potent lysosomal inhibitor; energetic impairment is required for WFA-induced apoptosis and growth inhibition and combining WFA and 2-DG is a promising therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Nethaji Muniraj
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sumit Siddharth
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arumugam Nagalingam
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa Walker
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juhyung Woo
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Balázs Győrffy
- MTA TTK Momentum Cancer Biomarker Research Group, Budapest, Hungary
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ed Gabrielson
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neeraj K Saxena
- Early Detection Research Group, National Cancer Institute, Rockville, MD, USA
| | - Dipali Sharma
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Xu S, Herschman HR. A Tumor Agnostic Therapeutic Strategy for Hexokinase 1-Null/Hexokinase 2-Positive Cancers. Cancer Res 2019; 79:5907-5914. [PMID: 31434645 DOI: 10.1158/0008-5472.can-19-1789] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/25/2019] [Accepted: 08/09/2019] [Indexed: 11/16/2022]
Abstract
Since Warburg's observation that most cancers exhibit elevated glycolysis, decades of research have attempted to reduce tumor glucose utilization as a therapeutic approach. Hexokinase (HK) activity is the first glycolytic enzymatic step; despite many attempts to inhibit HK activity, none has reached clinical application. Identification of HK isoforms, and recognition that most tissues express only HK1 while most tumors express HK1 and HK2, stimulated reducing HK2 activity as a therapeutic option. However, studies using HK2 shRNA and isogenic HK1+HK2- and HK1+HK2+ tumor cell pairs demonstrated that tumors expressing only HK1, while exhibiting reduced glucose consumption, progressed in vivo as well as tumors expressing both HK1 and HK2. However, HK1-HK2+ tumor subpopulations exist among many cancers. shRNA HK2 suppression in HK1-HK2+ liver cancer cells reduced xenograft tumor progression, in contrast to HK1+HK2+ cells. HK2 inhibition, and partial inhibition of both oxidative phosphorylation and fatty acid oxidation using HK2 shRNA and small-molecule drugs, prevented human liver HK1-HK2+ cancer xenograft progression. Using human multiple myeloma xenografts and mouse allogeneic models to identify potential clinical translational agents, triple therapies that include antisense HK2 oligonucleotides, metformin, and perhexiline prevent progression. These results suggest an agnostic approach for HK1-HK2+ cancers, regardless of tissue origin.
Collapse
Affiliation(s)
- Shili Xu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Harvey R Herschman
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California. .,Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Molecular Biology Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
30
|
Metformin in breast cancer: preclinical and clinical evidence. Curr Probl Cancer 2019; 44:100488. [PMID: 31235186 DOI: 10.1016/j.currproblcancer.2019.06.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/25/2019] [Accepted: 06/08/2019] [Indexed: 12/11/2022]
Abstract
Metformin, a well-acknowledged biguanide, safety profile and multiaction drug with low cost for management of type 2 diabetes, makes a first-class candidate for repurposing. The off-patent drug draws huge attention for repositioned for anticancer drug delivery recently. Still few unanswered questions are challenging, among them one leading question; can metformin use as a generic therapy for all breast cancer subtypes? And is metformin able to get over the problem of drug resistance? The review focused on the mechanisms of metformin action specifically for breast cancer therapy and overcoming the resistance; also discusses preclinical and ongoing and completed clinical trials. The existing limitation such as therapeutic dose specifically for cancer treatment, resistance of metformin in breast cancer and organic cation transporters heterogeneity of the drug opens up a new pathway for improved understanding and successful application as repurposed effective chemotherapeutics for breast cancer. However, much more additional research is needed to confirm the accurate efficacy of metformin treatment for prevention of cancer and its recurrence.
Collapse
|
31
|
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: 218] [Impact Index Per Article: 31.1] [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]
|