1
|
Pereira-Vieira J, Granja S, Celeiro SP, Barbosa-Matos C, Preto A, Queirós O, Ko YH, Casal M, Baltazar F. 3-Bromopyruvate boosts the effect of chemotherapy in acute myeloid leukemia by a pro-oxidant mechanism. Free Radic Biol Med 2025; 234:192-202. [PMID: 40222425 DOI: 10.1016/j.freeradbiomed.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Acute myeloid leukemia (AML) comprises a diverse group of blood cancers with varying genetic, phenotypic, and clinical traits, making development of targeted therapy challenging. Metabolic reprogramming in AML has been described as relevant for chemotherapy effectiveness. 3-Bromopyruvate (3-BP) is an anticancer agent that undermines energy metabolism of cancer cells. However, the effect of 3-BP in hematologic malignancies, such as AML, needs further investigation. Thus, we aimed to explore 3-BP as a chemo-sensitizing agent in AML. Different approaches of combining 3-BP with classical chemotherapy (daunorubicin and cytarabin) were tested in diverse AML cell lines. Cell sensitivity to the different drug combinations was analyzed by Trypan blue staining. The effect of pre-treatment with a non-toxic concentration of 3-BP was assessed on the AML cell metabolic profile (Western blot and immunofluorescence), mitochondrial activity (cytometry flow), and antioxidant capacity (colorimetric detection kit). KG-1 and MOLM13 cells showed increased sensitivity to chemotherapy (decreased EC50 values) after exposure to a non-toxic concentration (5 μM) of 3-BP. In both cell lines, 5 μM 3-BP decreased glucose consumption without changing extracellular lactate levels. 5 μM 3-BP treatment increased reactive oxygen species levels and decreased cell antioxidant capacity by depleting reduced glutathione levels in both KG-1 and MOLM13 cells. Our results demonstrate that non-toxic concentrations of 3-BP enhance the effect of classical chemotherapy in AML cells through a pro-oxidant mechanism. These data unveiled a new approach for AML treatment, using 3-BP or other pro-oxidant agents as co-adjuvants of chemotherapy, subsiding chemotherapy-induced side effects.
Collapse
Affiliation(s)
- Joana Pereira-Vieira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; REQUIMTE/LAQV, Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Department of Pathological, Cytological and Thanatological Anatomy, ESS|P.PORTO, 4200-072, Porto, Portugal.
| | - Sónia Pires Celeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Catarina Barbosa-Matos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Ana Preto
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057, Braga, Portugal; IBS-Institute of Science and Innovation for Bio-Sustainability, University of Minho, 4710-057, Braga, Portugal.
| | - Odília Queirós
- UNIPRO-Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116, Gandra, Portugal.
| | - Young Hee Ko
- KoDiscovery, LLC, Institute of Marine and Environmental Technology (IMET) Center, 701 East Pratt Street, Baltimore, MD, 21202, USA.
| | - Margarida Casal
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057, Braga, Portugal; IBS-Institute of Science and Innovation for Bio-Sustainability, University of Minho, 4710-057, Braga, Portugal.
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
2
|
Przywara K, Adamski R, Książczyk M, Suchodolski J, Cal M. 3-bromopyruvate induces morphological alteration and may initiate programmed cell death in Cryptococcus neoformans cells. Arch Microbiol 2024; 206:153. [PMID: 38472387 DOI: 10.1007/s00203-024-03894-9] [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: 12/04/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/14/2024]
Abstract
3-Bromopyruvate (3BP), known for its potent anticancer properties, also exhibits remarkable efficacy against the pathogenic fungus Cryptococcus neoformans. So far it has been proven that the main fungicidal activity of 3BP is based on ATP depletion and a reduction of intracellular level of glutathione. The presented study includes a broad range of methods to further investigate the mechanistic effects of 3BP on C. neoformans cells. The use of flow cytometry allowed a thorough examination of their survival during 3BP treatment, while observations using electron microscopy made it possible to note the changes in cellular morphology. Utilizing ruthenium red, the study suggests a mitochondrial pathway may initiate programmed cell death in response to 3BP. Analysis of free radical generation and gene expression changes supports this hypothesis. These findings enhance comprehension of 3BP's mechanisms in fungal cells, paving the way for its potential application as a therapeutic agent against cryptococcosis.
Collapse
Affiliation(s)
- Katarzyna Przywara
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Wrocław, Poland.
| | - Ryszard Adamski
- Laboratory of Microscopic Techniques, Faculty of Biological Sciences, University of Wrocław, Wrocław, Poland
| | - Marta Książczyk
- Department of Microbiology, Faculty of Biological Sciences, University of Wrocław, Wrocław, Poland
| | - Jakub Suchodolski
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Wrocław, Poland
| | - Magdalena Cal
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Wrocław, Poland
| |
Collapse
|
3
|
Marini HR, Facchini BA, di Francia R, Freni J, Puzzolo D, Montella L, Facchini G, Ottaiano A, Berretta M, Minutoli L. Glutathione: Lights and Shadows in Cancer Patients. Biomedicines 2023; 11:2226. [PMID: 37626722 PMCID: PMC10452337 DOI: 10.3390/biomedicines11082226] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
In cases of cellular injury, there is an observed increase in the production of reactive oxygen species (ROS). When this production becomes excessive, it can result in various conditions, including cancerogenesis. Glutathione (GSH), the most abundant thiol-containing antioxidant, is fundamental to re-establishing redox homeostasis. In order to evaluate the role of GSH and its antioxi-dant effects in patients affected by cancer, we performed a thorough search on Medline and EMBASE databases for relevant clinical and/or preclinical studies, with particular regard to diet, toxicities, and pharmacological processes. The conjugation of GSH with xenobiotics, including anti-cancer drugs, can result in either of two effects: xenobiotics may lose their harmful effects, or GSH conjugation may enhance their toxicity by inducing bioactivation. While being an interesting weapon against chemotherapy-induced toxicities, GSH may also have a potential protective role for cancer cells. New studies are necessary to better explain the relationship between GSH and cancer. Although self-prescribed glutathione (GSH) implementation is prevalent among cancer patients with the intention of reducing the toxic effects of anticancer treatments and potentially preventing damage to normal tissues, this belief lacks substantial scientific evidence for its efficacy in reducing toxicity, except in the case of cisplatin-related neurotoxicity. Therefore, the use of GSH should only be considered under medical supervision, taking into account the appropriate timing and setting.
Collapse
Affiliation(s)
- Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Bianca Arianna Facchini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80133 Napoli, Italy;
| | - Raffaele di Francia
- Gruppo Oncologico Ricercatori Italiani (GORI-ONLUS), 33170 Pordenone, Italy;
| | - José Freni
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Domenico Puzzolo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Liliana Montella
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Gaetano Facchini
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, 80131 Napoli, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| |
Collapse
|
4
|
Oudaert I, Van der Vreken A, Maes A, De Bruyne E, De Veirman K, Vanderkerken K, Menu E. Metabolic cross-talk within the bone marrow milieu: focus on multiple myeloma. Exp Hematol Oncol 2022; 11:49. [PMID: 36050788 PMCID: PMC9438316 DOI: 10.1186/s40164-022-00303-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer cells are well-known for their capacity to adapt their metabolism to their increasing energy demands which is necessary for tumor progression. This is no different for Multiple Myeloma (MM), a hematological cancer which develops in the bone marrow (BM), whereby the malignant plasma cells accumulate and impair normal BM functions. It has become clear that the hypoxic BM environment contributes to metabolic rewiring of the MM cells, including changes in metabolite levels, increased/decreased activity of metabolic enzymes and metabolic shifts. These adaptations will lead to a pro-tumoral environment stimulating MM growth and drug resistance In this review, we discuss the identified metabolic changes in MM and the BM microenvironment and summarize how these identified changes have been targeted (by inhibitors, genetic approaches or deprivation studies) in order to block MM progression and survival.
Collapse
Affiliation(s)
- Inge Oudaert
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Arne Van der Vreken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Anke Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium.
| |
Collapse
|
5
|
Potęga A. Glutathione-Mediated Conjugation of Anticancer Drugs: An Overview of Reaction Mechanisms and Biological Significance for Drug Detoxification and Bioactivation. Molecules 2022; 27:molecules27165252. [PMID: 36014491 PMCID: PMC9412641 DOI: 10.3390/molecules27165252] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022] Open
Abstract
The effectiveness of many anticancer drugs depends on the creation of specific metabolites that may alter their therapeutic or toxic properties. One significant route of biotransformation is a conjugation of electrophilic compounds with reduced glutathione, which can be non-enzymatic and/or catalyzed by glutathione-dependent enzymes. Glutathione usually combines with anticancer drugs and/or their metabolites to form more polar and water-soluble glutathione S-conjugates, readily excreted outside the body. In this regard, glutathione plays a role in detoxification, decreasing the likelihood that a xenobiotic will react with cellular targets. However, some drugs once transformed into thioethers are more active or toxic than the parent compound. Thus, glutathione conjugation may also lead to pharmacological or toxicological effects through bioactivation reactions. My purpose here is to provide a broad overview of the mechanisms of glutathione-mediated conjugation of anticancer drugs. Additionally, I discuss the biological importance of glutathione conjugation to anticancer drug detoxification and bioactivation pathways. I also consider the potential role of glutathione in the metabolism of unsymmetrical bisacridines, a novel prosperous class of anticancer compounds developed in our laboratory. The knowledge on glutathione-mediated conjugation of anticancer drugs presented in this review may be noteworthy for improving cancer therapy and preventing drug resistance in cancers.
Collapse
Affiliation(s)
- Agnieszka Potęga
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza Str. 11/12, 80-233 Gdańsk, Poland
| |
Collapse
|
6
|
Li J, Pan J, Liu Y, Luo X, Yang C, Xiao W, Li Q, Yang L, Zhang X. 3‑Bromopyruvic acid regulates glucose metabolism by targeting the c‑Myc/TXNIP axis and induces mitochondria‑mediated apoptosis in TNBC cells. Exp Ther Med 2022; 24:520. [PMID: 35837063 PMCID: PMC9257941 DOI: 10.3892/etm.2022.11447] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/01/2022] [Indexed: 12/03/2022] Open
Abstract
Aerobic glycolysis is commonly observed in tumor cells, including triple-negative breast cancer (TNBC) cells, and the rate of aerobic glycolysis is higher in TNBC cells than in non-TNBC cells. Hexokinase 2 (HK2) is a key enzyme in the glycolytic pathway and a target of the transcription factor c-Myc, which is highly expressed in TNBC and promotes aerobic glycolysis by enhancing HK2 expression. As an inhibitor of HK2, 3-bromopyruvic acid (3-BrPA) exhibits good therapeutic efficacy in intrahepatic and extrahepatic tumors and inhibits the proliferation of human tumor cells with high expression levels of c-Myc in vivo and in vitro. In addition, 3-BrPA combines with photodynamic therapy to inhibit TNBC cell migration. Thioredoxin-interacting protein (TXNIP) competes with c-Myc to reduce glucose consumption in tumor cells to restrain cell proliferation. A comparative analysis was performed in the present study in TNBC (HCC1143) and non-TNBC (MCF-7) cell lines to explore the effect of 3-BrPA on energy metabolism in TNBC cells and to investigate the possible mechanism of action. Cell viability and apoptosis were detected through Cell Counting Kit-8 and flow cytometry assays, respectively. Expression levels of HK2, glucose transporter 1, TXNIP, c-Myc and mitochondria-regulated apoptosis pathway proteins were measured through western blotting. 3-BrPA inhibited cell proliferation, downregulated c-Myc and HK2 expression, and upregulated TXNIP expression in TNBC cells, but it doesn't have the same effect on non-TNBC cells. Furthermore, 3-BrPA induced the typical manifestations of mitochondrial-mediated apoptosis such as decreasing Bcl-2 expression and increasing Bax, Cyt-C and Caspase-3 expression. The present results suggested that 3-BrPA promoted TXNIP protein expression and reduced HK2 expression in TNBC cells by downregulating c-Myc expression, inhibiting glycolysis including suppressing lactate generation, intracellular ATP generation and HK activity, inducing mitochondrial-mediated apoptosis and eventually suppressing TNBC cell proliferation. These findings may reveal a novel therapeutic target for the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jianmin Pan
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yang Liu
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaohui Luo
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Cheng Yang
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wangfa Xiao
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Qishang Li
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Lihui Yang
- Department of Nursing, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaodong Zhang
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
7
|
Cal M, Matyjaszczyk I, Filik K, Ogórek R, Ko Y, Ułaszewski S. Mitochondrial Function Are Disturbed in the Presence of the Anticancer Drug, 3-Bromopyruvate. Int J Mol Sci 2021; 22:ijms22126640. [PMID: 34205737 PMCID: PMC8235118 DOI: 10.3390/ijms22126640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
3-bromopuryvate (3-BP) is a compound with unique antitumor activity. It has a selective action against tumor cells that exhibit the Warburg effect. It has been proven that the action of 3-BP is pleiotropic: it acts on proteins, glycolytic enzymes, reduces the amount of ATP, induces the formation of ROS (reactive oxygen species), and induces nuclear DNA damage. Mitochondria are important organelles for the proper functioning of the cell. The production of cellular energy (ATP), the proper functioning of the respiratory chain, or participation in the production of amino acids are one of the many functions of mitochondria. Here, for the first time, we show on the yeast model that 3-BP acts in the eukaryotic cell also by influence on mitochondria and that agents inhibiting mitochondrial function can potentially be used in cancer therapy with 3-BP. We show that cells with functional mitochondria are more resistant to 3-BP than rho0 cells. Using an MTT assay (a colorimetric assay for assessing cell metabolic activity), we demonstrated that 3-BP decreased mitochondrial activity in yeast in a dose-dependent manner. 3-BP induces mitochondrial-dependent ROS generation which results in ∆sod2, ∆por1, or ∆gpx1 mutant sensitivity to 3-BP. Probably due to ROS mtDNA lesions rise during 3-BP treatment. Our findings may have a significant impact on the therapy with 3-BP.
Collapse
Affiliation(s)
- Magdalena Cal
- Department of Mycology and Genetics, University of Wroclaw, 51-148 Wroclaw, Poland; (I.M.); (R.O.); (S.U.)
- Correspondence: ; Tel.: +48-71-375-6269
| | - Irwin Matyjaszczyk
- Department of Mycology and Genetics, University of Wroclaw, 51-148 Wroclaw, Poland; (I.M.); (R.O.); (S.U.)
| | - Karolina Filik
- Laboratory of Medical Microbiology, Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Rafał Ogórek
- Department of Mycology and Genetics, University of Wroclaw, 51-148 Wroclaw, Poland; (I.M.); (R.O.); (S.U.)
| | - Young Ko
- KoDiscovery, LLC, Baltimore, MD 21202, USA;
| | - Stanisław Ułaszewski
- Department of Mycology and Genetics, University of Wroclaw, 51-148 Wroclaw, Poland; (I.M.); (R.O.); (S.U.)
| |
Collapse
|
8
|
Targeting Reactive Oxygen Species Metabolism to Induce Myeloma Cell Death. Cancers (Basel) 2021; 13:cancers13102411. [PMID: 34067602 PMCID: PMC8156203 DOI: 10.3390/cancers13102411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.
Collapse
|
9
|
Yi H, Shen X, Wang H, Luo S, Guo W, Chen P, Hu L, Liang L, Gong Y, Xiao X, Liu J. Protein mass spectrometry reveals lycorine exerting anti-multiple-myeloma effect by acting on VDAC2 and causing mitochondrial abnormalities. Biotechnol Lett 2021; 43:537-546. [PMID: 33386501 DOI: 10.1007/s10529-020-03053-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Two-dimensional electrophoresis (2-DE) and MALDI-TOF/TOF mass spectrometry were performed to compare the proteomic alterations of lycorine-treated and control cells to further investigate the anti-multiple myeloma (MM) mechanisms of lycorine. RESULTS Mass spectrometry results showed that after lycorine treatment of MM cells, 42% of the differentially expressed proteins had subcellular localization, mainly, on mitochondria. Voltage-dependent anion-selective channel protein 2 (VDAC2), the most abundant protein in the outer mitochondrial membrane, was up-regulated after treatment with lycorine and was subsequently verified by western blot analysis. Further studies on mitochondria found that lycorine was able to increase abnormal mitochondria and increase mitochondrial membrane potential. CONCLUSIONS Lycorine can achieve the effect of resisting multiple myeloma by acting on VDAC2 and causing mitochondrial abnormalities.
Collapse
Affiliation(s)
- Hui Yi
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Xiaokai Shen
- Xiangya Medical School, Central South University, Changsha, China
| | - Haiqin Wang
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Saiqun Luo
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Wancheng Guo
- Xiangya Medical School, Central South University, Changsha, China
| | - Peng Chen
- Xiangya Medical School, Central South University, Changsha, China
| | - Lei Hu
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Long Liang
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
| | - Yanfei Gong
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China.
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China.
| | - Jing Liu
- Molecular Biology Research Center & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
10
|
Wu S, Kuang H, Ke J, Pi M, Yang DH. Metabolic Reprogramming Induces Immune Cell Dysfunction in the Tumor Microenvironment of Multiple Myeloma. Front Oncol 2021; 10:591342. [PMID: 33520703 PMCID: PMC7845572 DOI: 10.3389/fonc.2020.591342] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor cells rewire metabolism to meet their increased nutritional demands, allowing the maintenance of tumor survival, proliferation, and expansion. Enhancement of glycolysis and glutaminolysis is identified in most, if not all cancers, including multiple myeloma (MM), which interacts with a hypoxic, acidic, and nutritionally deficient tumor microenvironment (TME). In this review, we discuss the metabolic changes including generation, depletion or accumulation of metabolites and signaling pathways, as well as their relationship with the TME in MM cells. Moreover, we describe the crosstalk among metabolism, TME, and changing function of immune cells during cancer progression. The overlapping metabolic phenotype between MM and immune cells is discussed. In this sense, targeting metabolism of MM cells is a promising therapeutic approach. We propose that it is important to define the metabolic signatures that may regulate the function of immune cells in TME in order to improve the response to immunotherapy.
Collapse
Affiliation(s)
- Shaojie Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huixian Kuang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Ke
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Medical Center of Assessment of Bone & Joint Diseases, Orthopaedic Hospital, General Hospital of Southern Theater Command, Guangzhou, China
| | - Manfei Pi
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| |
Collapse
|
11
|
Delphinidin Increases the Sensitivity of Ovarian Cancer Cell Lines to 3-Bromopyruvate. Int J Mol Sci 2021; 22:ijms22020709. [PMID: 33445795 PMCID: PMC7828231 DOI: 10.3390/ijms22020709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/17/2022] Open
Abstract
3-Bromopyruvic acid (3-BP) is a promising anticancer compound. Two ovary cancer (OC) cell lines, PEO1 and SKOV3, showed relatively high sensitivity to 3-BP (half maximal inhibitory concentration (IC50) of 18.7 and 40.5 µM, respectively). However, the further sensitization of OC cells to 3-BP would be desirable. Delphinidin (D) has been reported to be cytotoxic for cancer cell lines. We found that D was the most toxic for PEO1 and SKOV3 cells from among several flavonoids tested. The combined action of 3-BP and D was mostly synergistic in PEO1 cells and mostly weakly antagonistic in SKOV3 cells. The viability of MRC-5 fibroblasts was not affected by both compounds at concentrations of up to 100 µM. The combined action of 3-BP and D decreased the level of ATP and of dihydroethidium (DHE)-detectable reactive oxygen species (ROS), cellular mobility and cell staining with phalloidin and Mitotracker Red in both cell lines but increased the 2’,7’-dichlorofluorescein (DCFDA)-detectable ROS level and decreased the mitochondrial membrane potential and mitochondrial mass only in PEO1 cells. The glutathione level was increased by 3-BP+D only in SKOV3 cells. These differences may contribute to the lower sensitivity of SKOV3 cells to 3-BP+D. Our results point to the possibility of sensitization of at least some OC cells to 3-BP by D.
Collapse
|
12
|
Petricciuolo M, Davidescu M, Fettucciari K, Gatticchi L, Brancorsini S, Roberti R, Corazzi L, Macchioni L. The efficacy of the anticancer 3-bromopyruvate is potentiated by antimycin and menadione by unbalancing mitochondrial ROS production and disposal in U118 glioblastoma cells. Heliyon 2020; 6:e05741. [PMID: 33364504 PMCID: PMC7753915 DOI: 10.1016/j.heliyon.2020.e05741] [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: 08/25/2020] [Revised: 11/06/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming of tumour cells sustains cancer progression. Similar to other cancer cells, glioblastoma cells exhibit an increased glycolytic flow, which encourages the use of antiglycolytics as an effective complementary therapy. We used the antiglycolytic 3-bromopyruvate (3BP) as a metabolic modifier to treat U118 glioblastoma cells and investigated the toxic effects and the conditions to increase drug effectiveness at the lowest concentration. Cellular vitality was not affected by 3BP concentrations lower than 40 μM, although p-Akt dephosphorylation, p53 degradation, and ATP reduction occurred already at 30 μM 3BP. ROS generated in mitochondria were enhanced at 30 μM 3BP, possibly by unbalancing their generation and their disposal because of glutathione peroxidase inhibition. ROS triggered JNK and ERK phosphorylation, and cyt c release outside mitochondria, not accompanied by caspases-9 and -3 activation, probably due to 3BP-dependent alkylation of cysteine residues at caspase-9 catalytic site. To explore the possibility of sensitizing cells to 3BP treatment, we exploited 3BP effects on mitochondria by using 30 μM 3BP in association with antimycin A or menadione concentrations that in themselves exhibit poor toxicity. 3BP effect on cyt c release and cell vitality loss was potentiated due the greater oxidative stress induced by antimycin or menadione association with 3BP, supporting a preeminent role of mitochondrial ROS in 3BP toxicity. Indeed, the scavenger of mitochondrial superoxide MitoTEMPO counteracted 3BP-induced cyt c release and weakened the potentiating effect of 3BP/antimycin association. In conclusion, the biochemical mechanisms leading U118 glioblastoma cells to viability loss following 3BP treatment rely on mitochondrial ROS-dependent pathways. Their potentiation at low 3BP concentrations is consistent with the goal to minimize the toxic effect of the drug towards non-cancer cells.
Collapse
Affiliation(s)
- Maya Petricciuolo
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Magdalena Davidescu
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Katia Fettucciari
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Leonardo Gatticchi
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Stefano Brancorsini
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Rita Roberti
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Lanfranco Corazzi
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Lara Macchioni
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| |
Collapse
|
13
|
Oliveira GL, Coelho AR, Marques R, Oliveira PJ. Cancer cell metabolism: Rewiring the mitochondrial hub. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166016. [PMID: 33246010 DOI: 10.1016/j.bbadis.2020.166016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/12/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022]
Abstract
To adapt to tumoral environment conditions or even to escape chemotherapy, cells rapidly reprogram their metabolism to handle adversities and survive. Given the rapid rise of studies uncovering novel insights and therapeutic opportunities based on the role of mitochondria in tumor metabolic programing and therapeutics, this review summarizes most significant developments in the field. Taking in mind the key role of mitochondria on carcinogenesis and tumor progression due to their involvement on tumor plasticity, metabolic remodeling, and signaling re-wiring, those organelles are also potential therapeutic targets. Among other topics, we address the recent data intersecting mitochondria as of prognostic value and staging in cancer, by mitochondrial DNA (mtDNA) determination, and current inhibitors developments targeting mtDNA, OXPHOS machinery and metabolic pathways. We contribute for a holistic view of the role of mitochondria metabolism and directed therapeutics to understand tumor metabolism, to circumvent therapy resistance, and to control tumor development.
Collapse
Affiliation(s)
- Gabriela L Oliveira
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Ana R Coelho
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Ricardo Marques
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, UC-Biotech, University of Coimbra, Biocant Park, Cantanhede, Portugal.
| |
Collapse
|
14
|
The Anticancer Drug 3-Bromopyruvate Induces DNA Damage Potentially Through Reactive Oxygen Species in Yeast and in Human Cancer Cells. Cells 2020; 9:cells9051161. [PMID: 32397119 PMCID: PMC7290944 DOI: 10.3390/cells9051161] [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: 04/04/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
3-bromopyruvate (3-BP) is a small molecule with anticancer and antimicrobial activities. 3-BP is taken up selectively by cancer cells’ mono-carboxylate transporters (MCTs), which are highly overexpressed by many cancers. When 3-BP enters cancer cells it inactivates several glycolytic and mitochondrial enzymes, leading to ATP depletion and the generation of reactive oxygen species. While mechanisms of 3-BP uptake and its influence on cell metabolism are well understood, the impact of 3-BP at certain concentrations on DNA integrity has never been investigated in detail. Here we have collected several lines of evidence suggesting that 3-BP induces DNA damage probably as a result of ROS generation, in both yeast and human cancer cells, when its concentration is sufficiently low and most cells are still viable. We also demonstrate that in yeast 3-BP treatment leads to generation of DNA double-strand breaks only in S-phase of the cell cycle, possibly as a result of oxidative DNA damage. This leads to DNA damage, checkpoint activation and focal accumulation of the DNA response proteins. Interestingly, in human cancer cells exposure to 3-BP also induces DNA breaks that trigger H2A.X phosphorylation. Our current data shed new light on the mechanisms by which a sufficiently low concentration of 3-BP can induce cytotoxicity at the DNA level, a finding that might be important for the future design of anticancer therapies.
Collapse
|
15
|
D'Souza L, Bhattacharya D. Plasma cells: You are what you eat. Immunol Rev 2019; 288:161-177. [PMID: 30874356 DOI: 10.1111/imr.12732] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022]
Abstract
Plasma cells are terminally differentiated B lymphocytes that constitutively secrete antibodies. These antibodies can provide protection against pathogens, and their quantity and quality are the best clinical correlates of vaccine efficacy. As such, plasma cell lifespan is the primary determinant of the duration of humoral immunity. Yet dysregulation of plasma cell function can cause autoimmunity or multiple myeloma. The longevity of plasma cells is primarily dictated by nutrient uptake and non-transcriptionally regulated metabolic pathways. We have previously shown a positive effect of glucose uptake and catabolism on plasma cell longevity and function. In this review, we discuss these findings with an emphasis on nutrient uptake and its effects on respiratory capacity, lifespan, endoplasmic reticulum stress, and antibody secretion in plasma cells. We further discuss how some of these pathways may be dysregulated in multiple myeloma, potentially providing new therapeutic targets. Finally, we speculate on the connection between plasma cell intrinsic metabolism and systemic changes in nutrient availability and metabolic diseases.
Collapse
Affiliation(s)
- Lucas D'Souza
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
16
|
Dbouk NH, Covington MB, Nguyen K, Chandrasekaran S. Increase of reactive oxygen species contributes to growth inhibition by fluconazole in Cryptococcus neoformans. BMC Microbiol 2019; 19:243. [PMID: 31694529 PMCID: PMC6833255 DOI: 10.1186/s12866-019-1606-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/29/2019] [Indexed: 01/04/2023] Open
Abstract
Background Cryptococcus neoformans, a basidiomycetous yeast, is a fungal pathogen that can colonize the lungs of humans causing pneumonia and fungal meningitis in severely immunocompromised individuals. Recent studies have implied that the antifungal drug fluconazole (FLC) can induce oxidative stress in C. neoformans by increasing the production of reactive oxygen species (ROS), as presence of the antioxidant ascorbic acid (AA) could reverse the inhibitory effects of FLC on C. neoformans. However, in Candida albicans, AA has been shown to stimulate the expression of genes essential for ergosterol biosynthesis. Hence, the contribution of ROS in FLC-mediated growth inhibition remains unclear. Results In order to determine whether counteracting ROS generated by FLC in C. neoformans can contribute to diminishing inhibitory effects of FLC, we tested three other antioxidants in addition to AA, namely, pyrrolidine dithiocarbamate (PDTC), retinoic acid (RA), and glutathione (GSH). Our data confirm that there is an increase in ROS in the presence of FLC in C. neoformans. Importantly, all four antioxidants reversed FLC-mediated growth inhibition of C. neoformans to various extents. We further verified the involvement of increased ROS in FLC-mediated growth inhibition by determining that ROS-scavenging proteins, metallothioneins (CMT1 and CMT2), contribute to growth recovery by PDTC and AA during treatment with FLC. Conclusion Our study suggests that ROS contributes to FLC-mediated growth inhibition and points to a complex nature of antioxidant-mediated growth rescue in the presence of FLC.
Collapse
Affiliation(s)
| | | | - Kenny Nguyen
- Department of Biology, Furman University, Greenville, SC, USA
| | | |
Collapse
|
17
|
Niedźwiecka K, Ribas D, Casal M, Ułaszewski S. The Cryptococcus neoformans monocarboxylate transporter Jen4 is responsible for increased 3-bromopyruvate sensitivity. FEMS Yeast Res 2019; 19:5435460. [PMID: 30993332 DOI: 10.1093/femsyr/foz029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 04/06/2019] [Indexed: 12/13/2022] Open
Abstract
In the last decades, 3-bromopyruvate (3BP) has been intensively studied as a promising anticancer and antimicrobial agent. The transport of this drug inside the cell is a critical step for its toxicity in cancer and microorganisms. The Cryptococcus neoformans is the most sensitive species of microorganisms toward 3BP. Its cells exhibit the highest uptake rate of 3BP among all tested fungal strains. In Saccharomyces cerevisiae cells, the Jen1 transporter was found to be responsible for 3BP sensitivity. The deletion of Jen1 resulted in the abolishment of 3BP mediated transport. We functionally characterized the Jen4 protein, a Jen1 homologue of C. neoformans, and its role in the phenotypic 3BP sensitivity. The deletion of the CNAG_04704 gene, which encodes Jen4, was found to impair the mediated transport of 3BP and decrease 3BP sensitivity. Further heterologous expression of Jen4 in the S. cerevisiae jen1Δ ady2Δ strain restored the mediated transport of 3BP. The application of a green fluorescent protein fusion tag with the CNAG_04704, revealed the Jen4 labeled on the plasma membrane. The identification of 3BP transporters in pathogen cells is of great importance for understanding the mechanisms of 3BP action and to anticipate the application of this compound as an antimicrobial drug.
Collapse
Affiliation(s)
- Katarzyna Niedźwiecka
- Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland
| | - David Ribas
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Margarida Casal
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Stanisław Ułaszewski
- Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland
| |
Collapse
|
18
|
Brilhante RS, da Rocha MG, de Oliveira JS, España JDA, Pereira VS, Scm Castelo-Branco DD, A Pereira-Neto WD, Sidrim JJ, A Cordeiro RD, Rocha MF. Proton pump inhibitors versus Cryptococcus species: effects on in vitro susceptibility and melanin production. Future Microbiol 2019; 14:489-497. [PMID: 31033338 DOI: 10.2217/fmb-2018-0340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: This study aimed to evaluate the effects of proton pump inhibitors (PPIs) on growth and melanin production by Cryptococcus spp. Materials & methods: Minimum inhibitory concentrations (MICs) of omeprazole, esomeprazole, rabeprazole, pantoprazole and lansoprazole against Cryptococcus spp. were determined and the effect of PPIs on melanin production was evaluated, in the presence or absence of copper sulfate or glutathione. Results: PPIs showed MICs ranging from 125-1000 μg/ml and decreased melanization by Cryptococcus cells. Addition of copper sulfate or gluthatione restored melanogenesis of cells grown in the presence of PPIs. The presence of PPIs and glyphosate decreased copper sulfate toxicity (1 mM). Conclusion: PPIs inhibited melanogenesis of Cryptococcus spp., possibly by chelating copper or inhibiting copper ATPase transport.
Collapse
Affiliation(s)
- Raimunda Sn Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Maria G da Rocha
- Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará. Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| | - Jonathas S de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Jaime DA España
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Vandbergue S Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Débora de Scm Castelo-Branco
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Waldemiro de A Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - José Jc Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Rossana de A Cordeiro
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Marcos Fg Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology & Legal Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
- Postgraduate Program in Veterinary Sciences, College of Veterinary, State University of Ceará. Av. Dr. Silas Munguba, 1700, Campus do Itaperi, CEP: 60714-903, Fortaleza, Ceará, Brazil
| |
Collapse
|
19
|
Pichla M, Sroka J, Pienkowska N, Piwowarczyk K, Madeja Z, Bartosz G, Sadowska-Bartosz I. Metastatic prostate cancer cells are highly sensitive to 3-bromopyruvic acid. Life Sci 2019; 227:212-223. [PMID: 30928407 DOI: 10.1016/j.lfs.2019.03.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 01/06/2023]
Abstract
AIMS 3-Bromopyruvate (3-BP), an alkylating agent and a glycolytic inhibitor, is a promising anticancer agent, which can be efficient also against multidrug-resistant cancer cells. The aim of this study was to examine how 3-BP affects the survival and mobility of rat (MAT-LyLu and AT-2) and human (DU-145 and PC-3) metastatic prostate cancer cell lines. MAIN METHODS Cytotoxicity was estimated with Neutral Red. Cell mobility was analyzed by time-lapse microscopic monitoring of trajectories of individual cells at 5-min intervals for 6h. ATP was estimated with luciferin/luciferase and glutathione (GSH) with o-phthalaldehyde. Actin cytoskeleton was visualized with phalloidin conjugated with Atto-488. KEY FINDINGS All metastatic prostate cell lines studied were very sensitive to 3-BP (IC50 of 4-26μM). 3-Bromopyruvate drastically reduced cell movement even at concentrations of 5-10μM after 1h treatment. This compound depleted also cellular ATP and GSH, and disrupted actin cytoskeleton. SIGNIFICANCE The data obtained suggest that 3-BP can potentially be useful for treatment of metastatic prostate cancer and, especially, be efficient in limiting metastasis.
Collapse
Affiliation(s)
- Monika Pichla
- Department of Analytical Biochemistry, Faculty of Biology and Agriculture, University of Rzeszów, Zelwerowicza Street 4, 35-601 Rzeszów, Poland
| | - Jolanta Sroka
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Street 7, 30-387 Cracow, Poland
| | - Natalia Pienkowska
- Department of Analytical Biochemistry, Faculty of Biology and Agriculture, University of Rzeszów, Zelwerowicza Street 4, 35-601 Rzeszów, Poland
| | - Katarzyna Piwowarczyk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Street 7, 30-387 Cracow, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa Street 7, 30-387 Cracow, Poland
| | - Grzegorz Bartosz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, Pomorska Street 141/143, 90-236 Łódź, Poland
| | - Izabela Sadowska-Bartosz
- Department of Analytical Biochemistry, Faculty of Biology and Agriculture, University of Rzeszów, Zelwerowicza Street 4, 35-601 Rzeszów, Poland.
| |
Collapse
|
20
|
Fan T, Sun G, Sun X, Zhao L, Zhong R, Peng Y. Tumor Energy Metabolism and Potential of 3-Bromopyruvate as an Inhibitor of Aerobic Glycolysis: Implications in Tumor Treatment. Cancers (Basel) 2019; 11:317. [PMID: 30845728 PMCID: PMC6468516 DOI: 10.3390/cancers11030317] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor formation and growth depend on various biological metabolism processes that are distinctly different with normal tissues. Abnormal energy metabolism is one of the typical characteristics of tumors. It has been proven that most tumor cells highly rely on aerobic glycolysis to obtain energy rather than mitochondrial oxidative phosphorylation (OXPHOS) even in the presence of oxygen, a phenomenon called "Warburg effect". Thus, inhibition of aerobic glycolysis becomes an attractive strategy to specifically kill tumor cells, while normal cells remain unaffected. In recent years, a small molecule alkylating agent, 3-bromopyruvate (3-BrPA), being an effective glycolytic inhibitor, has shown great potential as a promising antitumor drug. Not only it targets glycolysis process, but also inhibits mitochondrial OXPHOS in tumor cells. Excellent antitumor effects of 3-BrPA were observed in cultured cells and tumor-bearing animal models. In this review, we described the energy metabolic pathways of tumor cells, mechanism of action and cellular targets of 3-BrPA, antitumor effects, and the underlying mechanism of 3-BrPA alone or in combination with other antitumor drugs (e.g., cisplatin, doxorubicin, daunorubicin, 5-fluorouracil, etc.) in vitro and in vivo. In addition, few human case studies of 3-BrPA were also involved. Finally, the novel chemotherapeutic strategies of 3-BrPA, including wafer, liposomal nanoparticle, aerosol, and conjugate formulations, were also discussed for future clinical application.
Collapse
Affiliation(s)
- Tengjiao Fan
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Xiaodong Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Yongzhen Peng
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment & Reuse Technology, Engineering Research Center of Beijing, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
21
|
Ko YH, Niedźwiecka K, Casal M, Pedersen PL, Ułaszewski S. 3-Bromopyruvate as a potent anticancer therapy in honor and memory of the late Professor André Goffeau. Yeast 2018; 36:211-221. [PMID: 30462852 DOI: 10.1002/yea.3367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 01/10/2023] Open
Abstract
3-Bromopyruvate (3BP) is a small, highly reactive molecule formed by bromination of pyruvate. In the year 2000, the antitumor properties of 3BP were discovered. Studies using animal models proved its high efficacy for anticancer therapy with no apparent side effects. This was also found to be the case in a limited number of cancer patients treated with 3BP. Due to the "Warburg effect," most tumor cells exhibit metabolic changes, for example, increased glucose consumption and lactic acid production resulting from mitochondrial-bound overexpressed hexokinase 2. Such alterations promote cell migration, immortality via inhibition of apoptosis, and less dependence on the availability of oxygen. Significantly, these attributes also make cancer cells more sensitive to agents, such as 3BP that inhibits energy production pathways without harming normal cells. This selectivity of 3BP is mainly due to overexpressed monocarboxylate transporters in cancer cells. Furthermore, 3BP is not a substrate for any pumps belonging to the ATP-binding cassette superfamily, which confers resistance to a variety of drugs. Also, 3BP has the capacity to induce multiple forms of cell death, by, for example, ATP depletion resulting from inactivation of both glycolytic and mitochondrial energy production pathways. In addition to its anticancer property, 3BP also exhibits antimicrobial activity. Various species of microorganisms are characterized by different susceptibility to 3BP inhibition. Among tested strains, the most sensitive was found to be the pathogenic yeast-like fungus Cryptococcus neoformans. Significantly, studies carried out in our laboratories have shown that 3BP exhibits a remarkable capacity to eradicate cancer cells, fungi, and algae.
Collapse
Affiliation(s)
- Young H Ko
- KoDiscovery, LLC, University of Maryland BioPark, Baltimore, Maryland, USA
| | | | - Margarida Casal
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Braga, Portugal
| | - Peter L Pedersen
- Department of Biological Chemistry and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
22
|
Jagielski T, Niedźwiecka K, Roeske K, Dyląg M. 3-Bromopyruvate as an Alternative Option for the Treatment of Protothecosis. Front Pharmacol 2018; 9:375. [PMID: 29725298 PMCID: PMC5917324 DOI: 10.3389/fphar.2018.00375] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/03/2018] [Indexed: 12/13/2022] Open
Abstract
Protothecosis is an unusual infection of both humans and animals caused by opportunistically pathogenic microalgae of the genus Prototheca. Until now, no standardized treatment protocols exist for the protothecal disease, boosted by a remarkable resistance of Prototheca spp. to a wide array of antimicrobial agents currently available in clinical use. Consequently, there is an urgent need for new effective drugs against Prototheca algae. In this study, the anti-Prototheca activity of 3-bromopyruvate (3BP), either alone or in combination with amphotericin B (AMB) was assessed in vitro, as well as the cytotoxicity of 3BP toward the bovine mammary epithelial cells and murine skin fibroblasts. The mean minimum inhibitory concentrations (MIC) and minimum algaecidal concentrations (MAC) were 0.85 ± 0.21 and 2.25 ± 0.54 mM for Prototheca wickerhamii, 1.25 ± 0.47 and 4.8 ± 1.03 mM for Prototheca blaschkeae, and 1.55 ± 0.69 and 5.6 ± 1.3 mM for Prototheca zopfii gen. 2, respectively. For all Prototheca strains tested, a synergistic interaction between 3BP and AMB was observed, resulting in about 4-fold reduction of their individual MICs, when used together. The elevated content of intracellular glutathione (GSH) was associated with a decreased susceptibility to 3BP. Both epithelial and fibroblast cells retained high viability upon treatment with 3BP at concentrations equivalent to the highest MIC recorded (3 mM) and 10-fold higher (30 mM), with the mean cell viability exceeding 80%, essentially the same as for the untreated cells. The results from these in vitro studies emphasize the high activity of 3BP against the Prototheca algae, its synergistic effect when used in combination with AMB, and the safety of the drug toward the tested mammalian cells. Along with the advantageous physico-chemical and pharmacokinetic properties, 3BP may be considered an effective and safe novel agent against the protothecal disease.
Collapse
Affiliation(s)
- Tomasz Jagielski
- Department of Applied Microbiology, Institute of Microbiology, University of Warsaw, Warsaw, Poland
| | - Katarzyna Niedźwiecka
- Department of Genetics, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Katarzyna Roeske
- Department of Applied Microbiology, Institute of Microbiology, University of Warsaw, Warsaw, Poland
| | - Mariusz Dyląg
- Department of Genetics, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
23
|
El Arfani C, De Veirman K, Maes K, De Bruyne E, Menu E. Metabolic Features of Multiple Myeloma. Int J Mol Sci 2018; 19:E1200. [PMID: 29662010 PMCID: PMC5979361 DOI: 10.3390/ijms19041200] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/07/2018] [Accepted: 04/10/2018] [Indexed: 01/19/2023] Open
Abstract
Cancer is known for its cellular changes contributing to tumour growth and cell proliferation. As part of these changes, metabolic rearrangements are identified in several cancers, including multiple myeloma (MM), which is a condition whereby malignant plasma cells accumulate in the bone marrow (BM). These metabolic changes consist of generation, inhibition and accumulation of metabolites and metabolic shifts in MM cells. Changes in the BM micro-environment could be the reason for such adjustments. Enhancement of glycolysis and glutaminolysis is found in MM cells compared to healthy cells. Metabolites and enzymes can be upregulated or downregulated and play a crucial role in drug resistance. Therefore, this review will focus on changes in glucose and glutamine metabolism linked with the emergence of drug resistance. Moreover, metabolites do not only affect other metabolic components to benefit cancer development; they also interfere with transcription factors involved in proliferation and apoptotic regulation.
Collapse
Affiliation(s)
- Chaima El Arfani
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium.
| |
Collapse
|
24
|
Dornfeld KJ, Skildum AJ. Mitochondria Remodeling in Cancer. MITOCHONDRIAL BIOLOGY AND EXPERIMENTAL THERAPEUTICS 2018:153-191. [DOI: 10.1007/978-3-319-73344-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
25
|
Indomethacin-based stimuli-responsive micelles combined with paclitaxel to overcome multidrug resistance. Oncotarget 2017; 8:111281-111294. [PMID: 29340053 PMCID: PMC5762321 DOI: 10.18632/oncotarget.22781] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/19/2017] [Indexed: 01/17/2023] Open
Abstract
Development of multidrug resistance against antitumor agents is a major limiting factor for the successful chemotherapy. Currently, both amphiphilic polymeric micelles and chemosensitizers have been proposed to overcome MDR during chemotherapy. Herein, the redox-responsive polymeric micelles composed of dextran and indomethacin (as chemosensitizer) using a disulfide bond as the linker are prepared (DEX-SS-IND) for delivery of antitumor agent paclitaxel (PTX). The high level of glutathione in tumor cells selectively breaks the disulfide bond, leading to the rapid breakdown and deformation of redox-responsive polymeric micelles. The data show that DEX-SS-IND can spontaneously form the stable micelles with high loading content (9.48 ± 0.41%), a favorable size of 45 nm with a narrow polydispersity (0.157), good stability, and glutathione-triggered drug release behavior due to the rapid breakdown of disulfide bond between DEX and IND. In vitro antitumor assay shows DEX-SS-IND/PTX micelles effectively inhibit the proliferation of PTX-resistant breast cancer (MCF-7/PTX) cells. More impressively, DEX-SS-IND/PTX micelles possess the improved plasma pharmacokinetics, enhanced antitumor efficacy on tumor growth in the xenograft models of MCF-7/PTX cells, and better in vivo safety. Overall, DEX-SS-IND/PTX micelles display a great potential for cancer treatment, especially for multidrug resistance tumors.
Collapse
|
26
|
Yadav S, Pandey SK, Kumar A, Kujur PK, Singh RP, Singh SM. Antitumor and chemosensitizing action of 3-bromopyruvate: Implication of deregulated metabolism. Chem Biol Interact 2017; 270:73-89. [DOI: 10.1016/j.cbi.2017.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/22/2023]
|
27
|
3-bromopyruvate and buthionine sulfoximine effectively kill anoikis-resistant hepatocellular carcinoma cells. PLoS One 2017; 12:e0174271. [PMID: 28362858 PMCID: PMC5376082 DOI: 10.1371/journal.pone.0174271] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/06/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND & AIMS Acquisition of anoikis resistance is a prerequisite for metastasis in hepatocellular carcinoma (HCC). However, little is known about how energy metabolism and antioxidant systems are altered in anoikis-resistant (AR) HCC cells. We evaluated anti-tumor effects of a combination treatment of 3-bromopyruvate (3-BP) and buthionine sulfoximine (BSO) in AR HCC cells. METHODS We compared glycolysis, reactive oxygen species (ROS) production, and chemoresistance among Huh-BAT, HepG2 HCC cells, and the corresponding AR cells. Expression of hexokinase II, gamma-glutamylcysteine synthetase (rGCS), and epithelial-mesenchymal transition (EMT) markers in AR cells was assessed. Anti-tumor effects of a combination treatment of 3-BP and BSO were evaluated in AR cells and an HCC xenograft mouse model. RESULTS AR HCC cells showed significantly higher chemoresistance, glycolysis and lower ROS production than attached cells. Expression of hexokinase II, rGCS, and EMT markers was higher in AR HCC cells than attached cells. A combination treatment of 3-BP/BSO effectively suppressed proliferation of AR HCC cells through apoptosis by blocking glycolysis and enhancing ROS levels. In xenograft mouse models, tumor growth derived from AR HCC cells was significantly suppressed in the group treated with 3-BP/BSO compared to the group treated with 3-BP or sorafenib. CONCLUSIONS These results demonstrated that a combination treatment of 3-BP/BSO had a synergistic anti-tumor effect in an AR HCC model. This strategy may be an effective adjuvant therapy for patients with sorafenib-resistant HCC.
Collapse
|
28
|
The HK2 Dependent "Warburg Effect" and Mitochondrial Oxidative Phosphorylation in Cancer: Targets for Effective Therapy with 3-Bromopyruvate. Molecules 2016; 21:molecules21121730. [PMID: 27983708 PMCID: PMC6273842 DOI: 10.3390/molecules21121730] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/09/2016] [Accepted: 12/11/2016] [Indexed: 12/30/2022] Open
Abstract
This review summarizes the current state of knowledge about the metabolism of cancer cells, especially with respect to the "Warburg" and "Crabtree" effects. This work also summarizes two key discoveries, one of which relates to hexokinase-2 (HK2), a major player in both the "Warburg effect" and cancer cell immortalization. The second discovery relates to the finding that cancer cells, unlike normal cells, derive as much as 60% of their ATP from glycolysis via the "Warburg effect", and the remaining 40% is derived from mitochondrial oxidative phosphorylation. Also described are selected anticancer agents which generally act as strong energy blockers inside cancer cells. Among them, much attention has focused on 3-bromopyruvate (3BP). This small alkylating compound targets both the "Warburg effect", i.e., elevated glycolysis even in the presence oxygen, as well as mitochondrial oxidative phosphorylation in cancer cells. Normal cells remain unharmed. 3BP rapidly kills cancer cells growing in tissue culture, eradicates tumors in animals, and prevents metastasis. In addition, properly formulated 3BP shows promise also as an effective anti-liver cancer agent in humans and is effective also toward cancers known as "multiple myeloma". Finally, 3BP has been shown to significantly extend the life of a human patient for which no other options were available. Thus, it can be stated that 3BP is a very promising new anti-cancer agent in the process of undergoing clinical development.
Collapse
|