1
|
Cordani M, Michetti F, Zarrabi A, Zarepour A, Rumio C, Strippoli R, Marcucci F. The role of glycolysis in tumorigenesis: From biological aspects to therapeutic opportunities. Neoplasia 2024; 58:101076. [PMID: 39476482 PMCID: PMC11555605 DOI: 10.1016/j.neo.2024.101076] [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: 08/02/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 11/11/2024]
Abstract
Glycolytic metabolism generates energy and intermediates for biomass production. Tumor-associated glycolysis is upregulated compared to normal tissues in response to tumor cell-autonomous or non-autonomous stimuli. The consequences of this upregulation are twofold. First, the metabolic effects of glycolysis become predominant over those mediated by oxidative metabolism. Second, overexpressed components of the glycolytic pathway (i.e. enzymes or metabolites) acquire new functions unrelated to their metabolic effects and which are referred to as "moonlighting" functions. These functions include induction of mutations and other tumor-initiating events, effects on cancer stem cells, induction of increased expression and/or activity of oncoproteins, epigenetic and transcriptional modifications, bypassing of senescence and induction of proliferation, promotion of DNA damage repair and prevention of DNA damage, antiapoptotic effects, inhibition of drug influx or increase of drug efflux. Upregulated metabolic functions and acquisition of new, non-metabolic functions lead to biological effects that support tumorigenesis: promotion of tumor initiation, stimulation of tumor cell proliferation and primary tumor growth, induction of epithelial-mesenchymal transition, autophagy and metastasis, immunosuppressive effects, induction of drug resistance and effects on tumor accessory cells. These effects have negative consequences on the prognosis of tumor patients. On these grounds, it does not come to surprise that tumor-associated glycolysis has become a target of interest in antitumor drug discovery. So far, however, clinical results with glycolysis inhibitors have fallen short of expectations. In this review we propose approaches that may allow to bypass some of the difficulties that have been encountered so far with the therapeutic use of glycolysis inhibitors.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy.
| |
Collapse
|
2
|
Chen C, Wang Z, Qin Y. Connections between metabolism and epigenetics: mechanisms and novel anti-cancer strategy. Front Pharmacol 2022; 13:935536. [PMID: 35935878 PMCID: PMC9354823 DOI: 10.3389/fphar.2022.935536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/29/2022] [Indexed: 12/26/2022] Open
Abstract
Cancer cells undergo metabolic adaptations to sustain their growth and proliferation under several stress conditions thereby displaying metabolic plasticity. Epigenetic modification is known to occur at the DNA, histone, and RNA level, which can alter chromatin state. For almost a century, our focus in cancer biology is dominated by oncogenic mutations. Until recently, the connection between metabolism and epigenetics in a reciprocal manner was spotlighted. Explicitly, several metabolites serve as substrates and co-factors of epigenetic enzymes to carry out post-translational modifications of DNA and histone. Genetic mutations in metabolic enzymes facilitate the production of oncometabolites that ultimately impact epigenetics. Numerous evidences also indicate epigenome is sensitive to cancer metabolism. Conversely, epigenetic dysfunction is certified to alter metabolic enzymes leading to tumorigenesis. Further, the bidirectional relationship between epigenetics and metabolism can impact directly and indirectly on immune microenvironment, which might create a new avenue for drug discovery. Here we summarize the effects of metabolism reprogramming on epigenetic modification, and vice versa; and the latest advances in targeting metabolism-epigenetic crosstalk. We also discuss the principles linking cancer metabolism, epigenetics and immunity, and seek optimal immunotherapy-based combinations.
Collapse
|
3
|
Kontaridis MI, Chennappan S. Mitochondria and the future of RASopathies: the emergence of bioenergetics. J Clin Invest 2022; 132:1-5. [PMID: 35426371 PMCID: PMC9017150 DOI: 10.1172/jci157560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RASopathies are a family of rare autosomal dominant disorders that affect the canonical Ras/MAPK signaling pathway and manifest as neurodevelopmental systemic syndromes, including Costello syndrome (CS). In this issue of the JCI, Dard et al. describe the molecular determinants of CS using a myriad of genetically modified models, including mice expressing HRAS p.G12S, patient-derived skin fibroblasts, hiPSC-derived human cardiomyocytes, an HRAS p.G12V zebrafish model, and human lentivirally induced fibroblasts overexpressing HRAS p.G12S or HRAS p.G12A. Mitochondrial proteostasis and oxidative phosphorylation were altered in CS, and inhibition of the AMPK signaling pathway mediated bioenergetic changes. Importantly, the pharmacological induction of this pathway restored cardiac function and reduced the developmental defects associated with CS. These findings identify a role for altered bioenergetics and provide insights into more effective treatment strategies for patients with RASopathies.
Collapse
Affiliation(s)
- Maria I. Kontaridis
- Masonic Medical Research Institute, Department of Biological Sciences and Translational Medicine, Utica, New York, USA
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Cardiology, Boston, Massachusetts, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Saravanakkumar Chennappan
- Masonic Medical Research Institute, Department of Biological Sciences and Translational Medicine, Utica, New York, USA
| |
Collapse
|
4
|
Abbott J, Mukherjee A, Wu W, Ye T, Jung HS, Cheung KM, Gertner RS, Basan M, Ham D, Park H. Multi-parametric functional imaging of cell cultures and tissues with a CMOS microelectrode array. LAB ON A CHIP 2022; 22:1286-1296. [PMID: 35266462 PMCID: PMC8963257 DOI: 10.1039/d1lc00878a] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 06/01/2023]
Abstract
Electrode-based impedance and electrochemical measurements can provide cell-biology information that is difficult to obtain using optical-microscopy techniques. Such electrical methods are non-invasive, label-free, and continuous, eliminating the need for fluorescence reporters and overcoming optical imaging's throughput/temporal resolution limitations. Nonetheless, electrode-based techniques have not been heavily employed because devices typically contain few electrodes per well, resulting in noisy aggregate readouts. Complementary metal-oxide-semiconductor (CMOS) microelectrode arrays (MEAs) have sometimes been used for electrophysiological measurements with thousands of electrodes per well at sub-cellular pitches, but only basic impedance mappings of cell attachment have been performed outside of electrophysiology. Here, we report on new field-based impedance mapping and electrochemical mapping/patterning techniques to expand CMOS-MEA cell-biology applications. The methods enable accurate measurement of cell attachment, growth/wound healing, cell-cell adhesion, metabolic state, and redox properties with single-cell spatial resolution (20 μm electrode pitch). These measurements allow the quantification of adhesion and metabolic differences of cells expressing oncogenes versus wild-type controls. The multi-parametric, cell-population statistics captured by the chip-scale integrated device opens up new avenues for fully electronic high-throughput live-cell assays for phenotypic screening and drug discovery applications.
Collapse
Affiliation(s)
- Jeffrey Abbott
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
- Department of Physics, Harvard University, Cambridge, Massachusetts, USA
| | - Avik Mukherjee
- Department of System Biology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Wenxuan Wu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Tianyang Ye
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Han Sae Jung
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Kevin M Cheung
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Rona S Gertner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
| | - Markus Basan
- Department of System Biology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Donhee Ham
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Hongkun Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.
- Department of Physics, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
5
|
Baryła M, Semeniuk-Wojtaś A, Róg L, Kraj L, Małyszko M, Stec R. Oncometabolites-A Link between Cancer Cells and Tumor Microenvironment. BIOLOGY 2022; 11:biology11020270. [PMID: 35205136 PMCID: PMC8869548 DOI: 10.3390/biology11020270] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment is the space between healthy tissues and cancer cells, created by the extracellular matrix, blood vessels, infiltrating cells such as immune cells, and cancer-associated fibroblasts. These components constantly interact and influence each other, enabling cancer cells to survive and develop in the host organism. Accumulated intermediate metabolites favoring dysregulation and compensatory responses in the cell, called oncometabolites, provide a method of communication between cells and might also play a role in cancer growth. Here, we describe the changes in metabolic pathways that lead to accumulation of intermediate metabolites: lactate, glutamate, fumarate, and succinate in the tumor and their impact on the tumor microenvironment. These oncometabolites are not only waste products, but also link all types of cells involved in tumor survival and progression. Oncometabolites play a particularly important role in neoangiogenesis and in the infiltration of immune cells in cancer. Oncometabolites are also associated with a disrupted DNA damage response and make the tumor microenvironment more favorable for cell migration. The knowledge summarized in this article will allow for a better understanding of associations between therapeutic targets and oncometabolites, as well as the direct effects of these particles on the formation of the tumor microenvironment. In the future, targeting oncometabolites could improve treatment standards or represent a novel method for fighting cancer.
Collapse
Affiliation(s)
- Maksymilian Baryła
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Aleksandra Semeniuk-Wojtaś
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
- Correspondence:
| | - Letycja Róg
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Maciej Małyszko
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| |
Collapse
|
6
|
Steroid receptor RNA activator gene footprint in the progression and drug resistance of colorectal cancer through oxidative phosphorylation pathway. Life Sci 2021; 285:119950. [PMID: 34520769 DOI: 10.1016/j.lfs.2021.119950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND The steroid receptor RNA activator 1 (SRA1) gene is involved in the progression of various cancers via different molecular mechanisms mediated by long non-coding RNA SRA (lncRNA SRA). This study aimed to evaluate the lncRNA SRA effect on the tumor progression of colorectal cancer (CRC). METHODS SRA1 expression was assessed in the cancer genome atlas datasets, CRC cell lines, and tumor specimens. Meta-analysis and gene co-expression network analysis were performed to identify pathways related to SRA1. RNA interference and cell treatment were utilized to examine the role of SRA1 expression in HT-29 and Caco-2 cell lines. Also, the effect of SRA1 expression was investigated on drug resistance, clinical parameters, and mutations in CRC samples. RESULTS The SRA1 transcripts, especially lncRNA SRA, were dysregulated in CRC tissue samples compared with normal tissue samples. Furthermore, SRA1 depletion decreased colony formation and proliferation while induced apoptosis in HT-29 and Caco-2 cells. In silico analyses indicated that SRA1 level was correlated with expression levels of oxidative phosphorylation (OXPHOS) genes. LncRNA SRA expression increased in response to the increased oxidative capacity, and when lncRNA SRA was knocked down, the expression level of OXPHOS pathway genes, including NDUFB5 and ATP5F1B, was changed. Also, KRAS-mutant samples had the highest SRA1 expression level. CONCLUSIONS LncRNA SRA could function as an oncogene through the OXPHOS pathway in CRC, and serve as a potential biomarker for identifying CRC subtype with KRAS mutations. The findings suggest that lncRNA SRA might be a therapeutic target to inhibit cell proliferation in CRC.
Collapse
|
7
|
Kamat V, Robbings BM, Jung SR, Kelly J, Hurley JB, Bube KP, Sweet IR. Fluidics system for resolving concentration-dependent effects of dissolved gases on tissue metabolism. eLife 2021; 10:e66716. [PMID: 34734803 PMCID: PMC8660022 DOI: 10.7554/elife.66716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Oxygen (O2) and other dissolved gases such as the gasotransmitters H2S, CO, and NO affect cell metabolism and function. To evaluate effects of dissolved gases on processes in tissue, we developed a fluidics system that controls dissolved gases while simultaneously measuring parameters of electron transport, metabolism, and secretory function. We use pancreatic islets, retina, and liver from rodents to highlight its ability to assess effects of O2 and H2S. Protocols aimed at emulating hypoxia-reperfusion conditions resolved a previously unrecognized transient spike in O2 consumption rate (OCR) following replenishment of O2, and tissue-specific recovery of OCR following hypoxia. The system revealed both inhibitory and stimulatory effects of H2S on insulin secretion rate from isolated islets. The unique ability of this new system to quantify metabolic state and cell function in response to precise changes in dissolved gases provides a powerful platform for cell physiologists to study a wide range of disease states.
Collapse
Affiliation(s)
- Varun Kamat
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | - Brian M Robbings
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Seung-Ryoung Jung
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| | | | - James B Hurley
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Kenneth P Bube
- Department of Mathematics, University of WashingtonSeattleUnited States
| | - Ian R Sweet
- University of Washington Medicine Diabetes Institute, University of WashingtonSeattleUnited States
| |
Collapse
|
8
|
Abstract
ATP is required for mammalian cells to remain viable and to perform genetically programmed functions. Maintenance of the ΔG′ATP hydrolysis of −56 kJ/mole is the endpoint of both genetic and metabolic processes required for life. Various anomalies in mitochondrial structure and function prevent maximal ATP synthesis through OxPhos in cancer cells. Little ATP synthesis would occur through glycolysis in cancer cells that express the dimeric form of pyruvate kinase M2. Mitochondrial substrate level phosphorylation (mSLP) in the glutamine-driven glutaminolysis pathway, substantiated by the succinate-CoA ligase reaction in the TCA cycle, can partially compensate for reduced ATP synthesis through both OxPhos and glycolysis. A protracted insufficiency of OxPhos coupled with elevated glycolysis and an auxiliary, fully operational mSLP, would cause a cell to enter its default state of unbridled proliferation with consequent dedifferentiation and apoptotic resistance, i.e., cancer. The simultaneous restriction of glucose and glutamine offers a therapeutic strategy for managing cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - Gabriel Arismendi-Morillo
- Electron Microscopy Laboratory, Biological Researches Institute, Faculty of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
9
|
Kariagina A, Lunt SY, McCormick JJ. Genomic and metabolomic analysis of step-wise malignant transformation in human skin fibroblasts. Carcinogenesis 2020; 41:656-665. [PMID: 31276576 DOI: 10.1093/carcin/bgz126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/24/2019] [Accepted: 07/03/2019] [Indexed: 12/28/2022] Open
Abstract
Metabolic changes accompanying a step-wise malignant transformation was investigated using a syngeneic lineage of human fibroblasts. Cell immortalization was associated with minor alterations in metabolism. Consecutive loss of cell cycle inhibition in immortalized cells resulted in increased levels of oxidative phosphorylation (OXPHOS). Overexpression of the H-Ras oncoprotein produced cells forming sarcomas in athymic mice. These transformed cells exhibited increased glucose consumption, glycolysis and a further increase in OXPHOS. Because of the markedly increased OXPHOS in transformed cells, the impact of a transaminase inhibitor, aminooxyacetic acid (AOA), which decreases glutamine influx to the tricarboxylic acid (TCA) cycle, was tested. Indeed, AOA significantly decreased proliferation of malignantly transformed fibroblasts and fibrosarcoma-derived cells in vitro and in vivo. AOA also decreased proliferation of cells susceptible to malignant transformation. Metabolomic studies in normal and transformed cells indicated that, in addition to the anticipated effect on the TCA cycle, AOA decreased production of nucleotides adenosine triphosphate (ATP) and uridine monophosphate. Exogenous nucleotides partially rescued decreased proliferation of the malignant cells treated with AOA. Our data indicate that AOA blocks several metabolic pathways essential for growth of malignant cells. Therefore, OXPHOS may provide important therapeutic targets for treatment of sarcoma.
Collapse
Affiliation(s)
- Anastasia Kariagina
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.,Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - J Justin McCormick
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA.,Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
10
|
Wilson RL, Connell JP, Grande-Allen KJ. Monitoring Oxygen Levels within Large, Tissue-Engineered Constructs Using Porphyin-Hydrogel Microparticles. ACS Biomater Sci Eng 2019; 5:4522-4530. [PMID: 33438417 DOI: 10.1021/acsbiomaterials.9b00257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A major barrier to the creation of engineered organs is the limited diffusion of oxygen through biological tissues. Advances in biofabrication bring us increasingly closer to complex vascular networks capable of supplying oxygen to large cellularized scaffolds. However, technologies for monitoring oxygen levels in engineered tissues do not accommodate imaging depths of more than a few dozen micrometers. Here, we report the creation of fluorescent porphyrin-hydrogel microparticles that can be used at depths of 2 mm into artificial tissues. By combining an oxygen-responsive porphyrin dye with a reference dye, the microparticles generate a ratiometric signal that is photostable, unaffected by attenuation from biological material, and responsive to physiological change in oxygen concentration. These microparticles can measure long-distance oxygen gradients within 3D, cellularized constructs and accurately report cellular oxygen consumption rates. Furthermore, they are compatible with a number of hydrogel polymerization chemistries and cell types, including primary human cells. We believe this technology will significantly advance efforts to visualize oxygen gradients in cellularized constructs and inform efforts to tissue engineer solid organs.
Collapse
Affiliation(s)
- Reid L Wilson
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States.,Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, United States
| | - Jennifer P Connell
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - K Jane Grande-Allen
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| |
Collapse
|
11
|
ER Ca 2+ release and store-operated Ca 2+ entry - partners in crime or independent actors in oncogenic transformation? Cell Calcium 2019; 82:102061. [PMID: 31394337 DOI: 10.1016/j.ceca.2019.102061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Ca2+ is a pleiotropic messenger that controls life and death decisions from fertilisation until death. Cellular Ca2+ handling mechanisms show plasticity and are remodelled throughout life to meet the changing needs of the cell. In turn, as the demands on a cell alter, for example through a change in its niche environment or its functional requirements, Ca2+ handling systems may be targeted to sustain the remodelled cellular state. Nowhere is this more apparent than in cancer. Oncogenic transformation is a multi-stage process during which normal cells become progressively differentiated towards a cancerous state that is principally associated with enhanced proliferation and avoidance of death. Ca2+ signalling is intimately involved in almost all aspects of the life of a transformed cell and alterations in Ca2+ handling have been observed in cancer. Moreover, this remodelling of Ca2+ signalling pathways is also required in some cases to sustain the transformed phenotype. As such, Ca2+ handling is hijacked by oncogenic processes to deliver and maintain the transformed phenotype. Central to generation of intracellular Ca2+ signals is the release of Ca2+ from the endoplasmic reticulum intracellular (ER) Ca2+ store via inositol 1,4,5-trisphosphate receptors (InsP3Rs). Upon depletion of ER Ca2+, store-operated Ca2+ entry (SOCE) across the plasma membrane occurs via STIM-gated Orai channels. SOCE serves to both replenish stores but also sustain Ca2+ signalling events. Here, we will discuss the role and regulation of these two signalling pathways and their interplay in oncogenic transformation.
Collapse
|
12
|
Mijanović O, Branković A, Panin AN, Savchuk S, Timashev P, Ulasov I, Lesniak MS. Cathepsin B: A sellsword of cancer progression. Cancer Lett 2019; 449:207-214. [PMID: 30796968 PMCID: PMC6488514 DOI: 10.1016/j.canlet.2019.02.035] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Clinical, biochemical and molecular biology studies have identified lysosome-encapsulated cellular proteases as critical risk factors for cancer progression. Cathepsins represent a group of such proteases aimed at maintenance of cellular homeostasis. Nevertheless, recent reports suggest that Cathepsin B executes other cellular programs such as controlling tumor growth, migration, invasion, angiogenesis, and metastases development. In fact, elevated levels of Cathepsins are found under different pathological conditions including inflammation, infection, neurodegenerative disease, and cancer. Furthermore, the discovery of Cathepsin B secretion and function as an extracellular matrix protein has broadened our appreciation for the impact of Cathepsin B on cancer progression. Underneath a façade of an intracellular protease with limited therapeutic potential hides a central role of cathepsins in extracellular functions. Moreover, this role is incredibly diverse from one condition to the next - from driving caspase-dependent apoptosis to facilitating tumor neovascularization and metastasis. Here we discuss the role of Cathepsin B in the oncogenic process and perspective the use of Cathepsin B for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Olja Mijanović
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ana Branković
- Department of Forensics, Academy of Criminalistic and Police Studies, Belgrade, Serbia
| | - Alexander N Panin
- Moscow State University of Food Production, 11, Volokolamskoe Shosse, Moscow, 125080, Russia
| | - Solomiia Savchuk
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA
| | - Peter Timashev
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Ilya Ulasov
- Institute of Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Maciej S Lesniak
- The University of Illinois at Chicago (UIC), Chicago, IL, USA; Northwestren University, Chicago, IL, 60611, USA.
| |
Collapse
|
13
|
Curcumin and its Potential for Systemic Targeting of Inflamm-Aging and Metabolic Reprogramming in Cancer. Int J Mol Sci 2019; 20:ijms20051180. [PMID: 30857125 PMCID: PMC6429141 DOI: 10.3390/ijms20051180] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 12/24/2022] Open
Abstract
Pleiotropic effects of curcumin have been the subject of intensive research. The interest in this molecule for preventive medicine may further increase because of its potential to modulate inflamm-aging. Although direct data related to its effect on inflamm-aging does not exist, there is a strong possibility that its well-known anti-inflammatory properties may be relevant to this phenomenon. Curcumin's binding to various proteins, which was shown to be dependent on cellular oxidative status, is yet another feature for exploration in depth. Finally, the binding of curcumin to various metabolic enzymes is crucial to curcumin's interference with powerful metabolic machinery, and can also be crucial for metabolic reprogramming of cancer cells. This review offers a synthesis and functional links that may better explain older data, some observational, in light of the most recent findings on curcumin. Our focus is on its modes of action that have the potential to alleviate specific morbidities of the 21st century.
Collapse
|
14
|
NCAM1 (CD56) promotes leukemogenesis and confers drug resistance in AML. Blood 2019; 133:2305-2319. [PMID: 30814062 DOI: 10.1182/blood-2018-12-889725] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Neural cell adhesion molecule 1 (NCAM1; CD56) is expressed in up to 20% of acute myeloid leukemia (AML) patients. NCAM1 is widely used as a marker of minimal residual disease; however, the biological function of NCAM1 in AML remains elusive. In this study, we investigated the impact of NCAM1 expression on leukemogenesis, drug resistance, and its role as a biomarker to guide therapy. Beside t(8;21) leukemia, NCAM1 expression was found in most molecular AML subgroups at highly heterogeneous expression levels. Using complementary genetic strategies, we demonstrated an essential role of NCAM1 in the regulation of cell survival and stress resistance. Perturbation of NCAM1 induced cell death or differentiation and sensitized leukemic blasts toward genotoxic agents in vitro and in vivo. Furthermore, Ncam1 was highly expressed in leukemic progenitor cells in a murine leukemia model, and genetic depletion of Ncam1 prolonged disease latency and significantly reduced leukemia-initiating cells upon serial transplantation. To further analyze the mechanism of the NCAM1-associated phenotype, we performed phosphoproteomics and transcriptomics in different AML cell lines. NCAM1 expression strongly associated with constitutive activation of the MAPK-signaling pathway, regulation of apoptosis, or glycolysis. Pharmacological inhibition of MEK1/2 specifically inhibited proliferation and sensitized NCAM1+ AML cells to chemotherapy. In summary, our data demonstrate that aberrant expression of NCAM1 is involved in the maintenance of leukemic stem cells and confers stress resistance, likely due to activation of the MAPK pathway. Targeting MEK1/2 sensitizes AML blasts to genotoxic agents, indicating a role for NCAM1 as a biomarker to guide AML treatment.
Collapse
|
15
|
Delic V, Noble K, Zivkovic S, Phan TA, Reynes C, Zhang Y, Phillips O, Claybaker C, Ta Y, Dinh VB, Cruz J, Prolla TA, Bradshaw PC. The effects of AICAR and rapamycin on mitochondrial function in immortalized mitochondrial DNA mutator murine embryonic fibroblasts. Biol Open 2018; 7:bio.033852. [PMID: 30177551 PMCID: PMC6262855 DOI: 10.1242/bio.033852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial DNA mutations accumulate with age and may play a role in stem cell aging as suggested by the premature aging phenotype of mitochondrial DNA polymerase gamma (POLG) exonuclease-deficient mice. Therefore, E1A immortalized murine embryonic fibroblasts (MEFs) from POLG exonuclease-deficient and wild-type (WT) mice were constructed. Surprisingly, when some E1A immortalized MEF lines were cultured in pyruvate-containing media they slowly became addicted to the pyruvate. The POLG exonuclease-deficient MEFs were more sensitive to several mitochondrial inhibitors and showed increased reactive oxygen species (ROS) production under standard conditions. When cultured in pyruvate-containing media, POLG exonuclease-deficient MEFs showed decreased oxygen consumption compared to controls. Increased AMP-activated protein kinase (AMPK) signaling and decreased mammalian target of rapamycin (mTOR) signaling delayed aging and influenced mitochondrial function. Therefore, the effects of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, or rapamycin, an mTOR inhibitor, on measures of mitochondrial function were determined. Rapamycin treatment transiently increased respiration only in WT MEFs and, under most conditions, increased ATP levels. Short term AICAR treatment transiently increased ROS production and, under most conditions, decreased ATP levels. Chronic AICAR treatment decreased respiration and ROS production in WT MEFs. These results demonstrate the context-dependent effects of AICAR and rapamycin on mitochondrial function. Summary: A novel mitochondrial DNA mutator murine embryonic fibroblast cell line was created and the effects of the anti-aging compounds rapamycin and AICAR on energy metabolism in these cells was determined.
Collapse
Affiliation(s)
- Vedad Delic
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama Birmingham School of Medicine, Birmingham, AL 35233, USA
| | - Kenyaria Noble
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Sandra Zivkovic
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Tam-Anh Phan
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Christian Reynes
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Yumeng Zhang
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA.,Department of Internal Medicine, University of South Florida, Tampa, FL 33606, USA
| | - Oluwakemi Phillips
- University of South Florida College of Medicine, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Charles Claybaker
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Yen Ta
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Vinh B Dinh
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Josean Cruz
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Tomas A Prolla
- Department of Genetics and Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Patrick C Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
16
|
Lehman JM, Hoeksema MD, Staub J, Qian J, Harris B, Callison JC, Miao J, Shi C, Eisenberg R, Chen H, Chen SC, Massion PP. Somatostatin receptor 2 signaling promotes growth and tumor survival in small-cell lung cancer. Int J Cancer 2018; 144:1104-1114. [PMID: 30152518 DOI: 10.1002/ijc.31771] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Abstract
Somatostatin receptor 2 (SSTR2) is overexpressed in a majority of neuroendocrine neoplasms, including small-cell lung carcinomas (SCLCs). SSTR2 was previously considered an inhibitory receptor on cell growth, but its agonists had poor clinical responses in multiple clinical trials. The role of this receptor as a potential therapeutic target in lung cancer merits further investigation. We evaluated the expression of SSTR2 in a cohort of 96 primary tumors from patients with SCLC and found 48% expressed SSTR2. Correlation analysis in both CCLE and an SCLC RNAseq cohort confirmed high-level expression and identified an association between NEUROD1 and SSTR2. There was a significant association with SSTR2 expression profile and poor clinical outcome. We tested whether SSTR2 expression might contribute to tumor progression through activation of downstream signaling pathways, using in vitro and in vivo systems and downregulated SSTR2 expression in lung cancer cells by shRNA. SSTR2 downregulation led to increased apoptosis and dramatically decreased tumor growth in vitro and in vivo in multiple cell lines with decreased AMPKα phosphorylation and increased oxidative metabolism. These results demonstrate a role for SSTR2 signaling in SCLC and suggest that SSTR2 is a poor prognostic biomarker in SCLC and potential future therapeutic signaling target.
Collapse
Affiliation(s)
- Jonathan M Lehman
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Megan D Hoeksema
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jeremy Staub
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Bradford Harris
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - J Clay Callison
- University of Tennessee Graduate School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Jennifer Miao
- Vanderbilt University School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Chanjuan Shi
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Rosana Eisenberg
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Heidi Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Sheau-Chiann Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Pierre P Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| |
Collapse
|
17
|
Dard L, Bellance N, Lacombe D, Rossignol R. RAS signalling in energy metabolism and rare human diseases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:845-867. [PMID: 29750912 DOI: 10.1016/j.bbabio.2018.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/12/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
The RAS pathway is a highly conserved cascade of protein-protein interactions and phosphorylation that is at the heart of signalling networks that govern proliferation, differentiation and cell survival. Recent findings indicate that the RAS pathway plays a role in the regulation of energy metabolism via the control of mitochondrial form and function but little is known on the participation of this effect in RAS-related rare human genetic diseases. Germline mutations that hyperactivate the RAS pathway have been discovered and linked to human developmental disorders that are known as RASopathies. Individuals with RASopathies, which are estimated to affect approximately 1/1000 human birth, share many overlapping characteristics, including cardiac malformations, short stature, neurocognitive impairment, craniofacial dysmorphy, cutaneous, musculoskeletal, and ocular abnormalities, hypotonia and a predisposition to developing cancer. Since the identification of the first RASopathy, type 1 neurofibromatosis (NF1), which is caused by the inactivation of neurofibromin 1, several other syndromes have been associated with mutations in the core components of the RAS-MAPK pathway. These syndromes include Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NSML), which was formerly called LEOPARD syndrome, Costello syndrome (CS), cardio-facio-cutaneous syndrome (CFC), Legius syndrome (LS) and capillary malformation-arteriovenous malformation syndrome (CM-AVM). Here, we review current knowledge about the bioenergetics of the RASopathies and discuss the molecular control of energy homeostasis and mitochondrial physiology by the RAS pathway.
Collapse
Affiliation(s)
- L Dard
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France
| | - N Bellance
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France; CHU de Bordeaux, Service de Génétique Médicale, F-33076 Bordeaux, France
| | - R Rossignol
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France.
| |
Collapse
|
18
|
Alsady M, de Groot T, Kortenoeven MLA, Carmone C, Neijman K, Bekkenkamp-Grovenstein M, Engelke U, Wevers R, Baumgarten R, Korstanje R, Deen PMT. Lithium induces aerobic glycolysis and glutaminolysis in collecting duct principal cells. Am J Physiol Renal Physiol 2017; 314:F230-F239. [PMID: 29070571 DOI: 10.1152/ajprenal.00297.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Lithium, given to bipolar disorder patients, causes nephrogenic diabetes insipidus (Li-NDI), a urinary-concentrating defect. Li-NDI occurs due to downregulation of principal cell AQP2 expression, which coincides with principal cell proliferation. The metabolic effect of lithium on principal cells, however, is unknown and investigated here. In earlier studies, we showed that the carbonic anhydrase (CA) inhibitor acetazolamide attenuated Li-induced downregulation in mouse-collecting duct (mpkCCD) cells. Of the eight CAs present in mpkCCD cells, siRNA and drug treatments showed that downregulation of CA9 and to some extent CA12 attenuated Li-induced AQP2 downregulation. Moreover, lithium induced cell proliferation and increased the secretion of lactate. Lithium also increased urinary lactate levels in wild-type mice that developed Li-NDI but not in lithium-treated mice lacking ENaC, the principal cell entry site for lithium. Inhibition of aerobic glycolysis with 2-deoxyglucose (2DG) attenuated lithium-induced AQP2 downregulation in mpkCCD cells but did not attenuate Li-NDI in mice. Interestingly, NMR analysis demonstrated that lithium also increased the urinary succinate, fumarate, citrate, and NH4+ levels, which were, in contrast to lactate, not decreased by 2DG. Together, our data reveal that lithium induces aerobic glycolysis and glutaminolysis in principal cells and that inhibition of aerobic glycolysis, but not the glutaminolysis, does not attenuate Li-NDI.
Collapse
Affiliation(s)
- Mohammad Alsady
- Department of Physiology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Theun de Groot
- Department of Physiology, Radboud University Medical Center , Nijmegen , The Netherlands.,The Jackson Laboratory, Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory , Bar Harbor, Maine
| | | | - Claudia Carmone
- Department of Physiology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Kim Neijman
- Department of Physiology, Radboud University Medical Center , Nijmegen , The Netherlands
| | | | - Udo Engelke
- Department of Laboratory Medicine, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Ron Wevers
- Department of Laboratory Medicine, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Ruben Baumgarten
- Society of Experimental Laboratory Medicine , Amersfoort , The Netherlands
| | - Ron Korstanje
- The Jackson Laboratory, Nathan Shock Center of Excellence in the Basic Biology of Aging, The Jackson Laboratory , Bar Harbor, Maine
| | | |
Collapse
|
19
|
Seyfried TN, Yu G, Maroon JC, D'Agostino DP. Press-pulse: a novel therapeutic strategy for the metabolic management of cancer. Nutr Metab (Lond) 2017; 14:19. [PMID: 28250801 PMCID: PMC5324220 DOI: 10.1186/s12986-017-0178-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A shift from respiration to fermentation is a common metabolic hallmark of cancer cells. As a result, glucose and glutamine become the prime fuels for driving the dysregulated growth of tumors. The simultaneous occurrence of "Press-Pulse" disturbances was considered the mechanism responsible for reduction of organic populations during prior evolutionary epochs. Press disturbances produce chronic stress, while pulse disturbances produce acute stress on populations. It was only when both disturbances coincide that population reduction occurred. METHODS This general concept can be applied to the management of cancer by creating chronic metabolic stresses on tumor cell energy metabolism (press disturbance) that are coupled to a series of acute metabolic stressors that restrict glucose and glutamine availability while also stimulating cancer-specific oxidative stress (pulse disturbances). The elevation of non-fermentable ketone bodies protect normal cells from energy stress while further enhancing energy stress in tumor cells that lack the metabolic flexibility to use ketones as an efficient energy source. Mitochondrial abnormalities and genetic mutations make tumor cells vulnerable metabolic stress. RESULTS The press-pulse therapeutic strategy for cancer management is illustrated with calorie restricted ketogenic diets (KD-R) used together with drugs and procedures that create both chronic and intermittent acute stress on tumor cell energy metabolism, while protecting and enhancing the energy metabolism of normal cells. CONCLUSIONS Optimization of dosing, timing, and scheduling of the press-pulse therapeutic strategy will facilitate the eradication of tumor cells with minimal patient toxicity. This therapeutic strategy can be used as a framework for the design of clinical trials for the non-toxic management of most cancers.
Collapse
Affiliation(s)
| | - George Yu
- George Washington University Medical Center Washington DC, and Aegis Medical & Research Associates Annapolis, Maryland, USA
| | - Joseph C Maroon
- Department of Neurosurgery, University of Pittsburgh Medical Center, Suite 5C, 200 Lothrop St, Pittsburgh, PA USA
| | - Dominic P D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida USA
| |
Collapse
|
20
|
Hong SE, Kim CS, An S, Kim HA, Hwang SG, Song JY, Lee JK, Hong J, Kim JI, Noh WC, Jin HO, Park IC. TRAIL restores DCA/metformin-mediated cell death in hypoxia. Biochem Biophys Res Commun 2016; 478:1389-95. [DOI: 10.1016/j.bbrc.2016.08.134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 08/24/2016] [Indexed: 01/01/2023]
|
21
|
Salazar-Ramiro A, Ramírez-Ortega D, Pérez de la Cruz V, Hérnandez-Pedro NY, González-Esquivel DF, Sotelo J, Pineda B. Role of Redox Status in Development of Glioblastoma. Front Immunol 2016; 7:156. [PMID: 27199982 PMCID: PMC4844613 DOI: 10.3389/fimmu.2016.00156] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/11/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive neoplasia, prognosis remains dismal, and current therapy is mostly palliative. There are no known risk factors associated with gliomagenesis; however, it is well established that chronic inflammation in brain tissue induces oxidative stress in astrocytes and microglia. High quantities of reactive species of oxygen into the cells can react with several macromolecules, including chromosomal and mitochondrial DNA, leading to damage and malfunction of DNA repair enzymes. These changes bring genetic instability and abnormal metabolic processes, favoring oxidative environment and increase rate of cell proliferation. In GBM, a high metabolic rate and increased basal levels of reactive oxygen species play an important role as chemical mediators in the regulation of signal transduction, protecting malignant cells from apoptosis, thus creating an immunosuppressive environment. New redox therapeutics could reduce oxidative stress preventing cellular damage and high mutation rate accompanied by chromosomal instability, reducing the immunosuppressive environment. In addition, therapies directed to modulate redox rate reduce resistance and moderate the high rate of cell proliferation, favoring apoptosis of tumoral cells. This review describes the redox status in GBM, and how this imbalance could promote gliomagenesis through genomic and mitochondrial DNA damage, inducing the pro-oxidant and proinflammatory environment involved in tumor cell proliferation, resistance, and immune escape. In addition, some therapeutic agents that modulate redox status and might be advantageous in therapy against GBM are described.
Collapse
Affiliation(s)
- Aleli Salazar-Ramiro
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Daniela Ramírez-Ortega
- Neurochemistry Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | | | | | | | - Julio Sotelo
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| | - Benjamín Pineda
- Neuroimmunology and Neuro-Oncology Unit, National Neurology and Neurosurgery Institute (INNN) , Mexico City , Mexico
| |
Collapse
|
22
|
Madhu B, Narita M, Jauhiainen A, Menon S, Stubbs M, Tavaré S, Narita M, Griffiths JR. Metabolomic changes during cellular transformation monitored by metabolite-metabolite correlation analysis and correlated with gene expression. Metabolomics 2015; 11:1848-1863. [PMID: 26491426 PMCID: PMC4605990 DOI: 10.1007/s11306-015-0838-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022]
Abstract
To investigate metabolic changes during cellular transformation, we used a 1H NMR based metabolite-metabolite correlation analysis (MMCA) method, which permits analysis of homeostatic mechanisms in cells at the steady state, in an inducible cell transformation model. Transcriptomic data were used to further explain the results. Transformed cells showed many more metabolite-metabolite correlations than control cells. Some had intuitively plausible explanations: a shift from glycolysis to amino acid oxidation after transformation was accompanied by a strongly positive correlation between glucose and glutamine and a strongly negative one between lactate and glutamate; there were also many correlations between the branched chain amino acids and the aromatic amino acids. Others remain puzzling: after transformation strong positive correlations developed between choline and a group of five amino acids, whereas the same amino acids showed negative correlations with phosphocholine, a membrane phospholipid precursor. MMCA in conjunction with transcriptome analysis has opened a new window into the metabolome.
Collapse
Affiliation(s)
- Basetti Madhu
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - Masako Narita
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - Alexandra Jauhiainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Early Clinical Biometrics, AstraZeneca AB R&D, Mölndal, Sweden
| | - Suraj Menon
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - Marion Stubbs
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - Simon Tavaré
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - Masashi Narita
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| | - John R. Griffiths
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE UK
| |
Collapse
|
23
|
How do changes in the mtDNA and mitochondrial dysfunction influence cancer and cancer therapy? Challenges, opportunities and models. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 764:16-30. [DOI: 10.1016/j.mrrev.2015.01.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
|
24
|
Lee JE, Jo DE, Lee AJ, Park HK, Youn K, Yun EY, Hwang JS, Jun M, Kang BH. Hepatoprotective and Anticancer Activities of Allomyrina dichotoma Larvae. ACTA ACUST UNITED AC 2015. [DOI: 10.5352/jls.2015.25.3.307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
25
|
Shen H, Decollogne S, Dilda PJ, Hau E, Chung SA, Luk PP, Hogg PJ, McDonald KL. Dual-targeting of aberrant glucose metabolism in glioblastoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:14. [PMID: 25652202 PMCID: PMC4324653 DOI: 10.1186/s13046-015-0130-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/28/2015] [Indexed: 01/02/2023]
Abstract
Background Glioblastoma (GBM) is the most common and malignant primary brain tumor. In contrast to some other tumor types, aberrant glucose metabolism is an important component of GBM growth and chemoresistance. Recent studies of human orthotopic GBM in mice and in situ demonstrated GBM cells rely on both glycolysis and mitochondrial oxidation for glucose catabolism. These observations suggest that the homeostasis of energy metabolism of GBM cells might be further disturbed by dual-inhibition of glucose metabolism. The present study aimed to evaluate the efficacy and the mechanisms of dual-targeting therapy in GBM cells. Methods Representative GBM cells (immortalized GBM cell lines and patient-derived GBM cells) and non-cancerous cells were treated with 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid (PENAO), an in-house designed novel arsenic-based mitochondrial toxin, in combination with dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor. The efficacy of this combinatorial therapy was evaluated by MTS assay, clonogenic surviving assay and apoptotic assays. The underlying mechanisms of this dual-targeting treatment were unraveled by using mitochondrial membrane potential measurements, cytosol/mitochondrial ROS detection, western blotting, extracellular flux assay and mass spectrometry. Results As monotherapies, both PENAO and DCA induced proliferation arrest in a panel of GBM cell lines and primary isolates. PENAO inhibited oxygen consumption, induced oxidative stress and depolarized mitochondrial membrane potential, which in turn activated mitochondria-mediated apoptosis. By combining DCA with PENAO, the two drugs worked synergistically to inhibit cell proliferation (but had no significant effect on non-cancerous cells), impair the clonogenicity, and induce mitochondria-mediated apoptosis. An oxidative stress of mitochondrial origin takes a prominent place in the mechanism by which the combination of PENAO and DCA induces cell death. Additionally, PENAO-induced oxidative damage was enhanced by DCA through glycolytic inhibition which in turn diminished acid production induced by PENAO. Moreover, DCA treatment also led to an alteration in the multidrug resistance (MDR) phenotype of GBM cells, thereby leading to an increased cytosolic accumulation of PENAO. Conclusions The findings of this study shed a new light with respect to the dual-targeting of glucose metabolism in GBM cells and the innovative combination of PENAO and DCA shows promise in expanding GBM therapies.
Collapse
Affiliation(s)
- Han Shen
- Cure Brain Cancer Neuro-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, 2052, Australia.
| | - Stephanie Decollogne
- Tumour Metabolism Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Pierre J Dilda
- Tumour Metabolism Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Eric Hau
- Cure Brain Cancer Neuro-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, 2052, Australia. .,Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia.
| | - Sylvia A Chung
- Cure Brain Cancer Neuro-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, 2052, Australia.
| | - Peter P Luk
- Tumour Metabolism Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Philip J Hogg
- Tumour Metabolism Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Kerrie L McDonald
- Cure Brain Cancer Neuro-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, 2052, Australia.
| |
Collapse
|
26
|
Obre E, Rossignol R. Emerging concepts in bioenergetics and cancer research: Metabolic flexibility, coupling, symbiosis, switch, oxidative tumors, metabolic remodeling, signaling and bioenergetic therapy. Int J Biochem Cell Biol 2015; 59:167-81. [DOI: 10.1016/j.biocel.2014.12.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 12/13/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023]
|
27
|
Deletion of Pim kinases elevates the cellular levels of reactive oxygen species and sensitizes to K-Ras-induced cell killing. Oncogene 2014; 34:3728-36. [PMID: 25241892 PMCID: PMC4369476 DOI: 10.1038/onc.2014.306] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/05/2014] [Accepted: 08/13/2014] [Indexed: 02/08/2023]
Abstract
The Pim protein kinases contribute to transformation by enhancing the activity of oncogenic Myc and Ras, which drives significant metabolic changes during tumorigenesis. In this report, we demonstrate that mouse embryo fibroblasts (MEFs) lacking all three isoforms of Pim protein kinases, triple knockout (TKO), cannot tolerate the expression of activated K-Ras (K-RasG12V) and undergo cell death. Transduction of K-RasG12V into these cells markedly increased the level of cellular reactive oxygen species (ROS). The addition of N-acetyl cysteine attenuates ROS production and reversed the cytotoxic effects of K-RasG12V in the TKO MEFs. The altered cellular redox state caused by the loss of Pim occurred as a result of lower levels of metabolic intermediates in the glycolytic and pentose phosphate pathways as well as abnormal mitochondrial oxidative phosphorylation. TKO MEFs exhibit reduced levels of superoxide dismutase (Sod), glutathione peroxidase 4 (Gpx4) and peroxiredoxin 3 (Prdx3) that render them susceptible to killing by K-RasG12V-mediated ROS production. In contrast, the transduction of c-Myc into TKO cells can overcome the lack of Pim protein kinases by regulating cellular metabolism and Sod2. In the absence of the Pim kinases, c-Myc transduction permitted K-RasG12V-induced cell growth by decreasing Ras-induced cellular ROS levels. These results demonstrate that the Pim protein kinases play an important role in regulating cellular redox, metabolism and K-Ras-stimulated cell growth.
Collapse
|
28
|
Understanding the pharmacological properties of a metabolic PET tracer in prostate cancer. Proc Natl Acad Sci U S A 2014; 111:7254-9. [PMID: 24785505 DOI: 10.1073/pnas.1405240111] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Generally, solid tumors (>400 mm(3)) are inherently acidic, with more aggressive growth producing greater acidity. If the acidity could be targeted as a biomarker, it would provide a means to gauge the pace of tumor growth and degree of invasiveness, as well as providing a basis for predicting responses to pH-dependent chemotherapies. We have developed a (64)Cu pH (low) insertion peptide (pHLIP) for targeting, imaging, and quantifying acidic tumors by PET, and our findings reveal utility in assessing prostate tumors. The new pHLIP version limits indiscriminate healthy tissue binding, and we demonstrate its targeting of extracellular acidification in three different prostate cancer models, each with different vascularization and acid-extruding protein carbonic anhydrase IX (CAIX) expression. We then describe the tumor distribution of this radiotracer ex vivo, in association with blood perfusion and known biomarkers of acidity, such as hypoxia, lactate dehydrogenase A, and CAIX. We find that the probe reveals metabolic variations between and within tumors, and discriminates between necrotic and living tumor areas.
Collapse
|
29
|
Mitochondria-targeted antioxidant and glycolysis inhibition: synergistic therapy in hepatocellular carcinoma. Anticancer Drugs 2014; 24:881-8. [PMID: 23872912 DOI: 10.1097/cad.0b013e32836442c6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Mito-carboxy proxyl (Mito-CP), a lipophilic cationic nitroxide, accumulates in the mitochondria because of the large negative transmembrane potential. Studies have shown that these agents act by disrupting the energy-producing mechanism, inducing mitochondrial-mediated apoptosis, and also enhancing the action of other chemotherapeutic agents in cancer cells. We hypothesized that the combination of Mito-CP and glycolysis inhibitor, 2-deoxyglucose (2-DG), would synergistically inhibit HCC in vitro. HepG2 cells and primary hepatocytes were treated with various combinations of Mito-CP and 2-DG. Cell cytotoxicity was measured using the methylthiazolyldiphenyl-tetrazolium bromide assay and ATP bioluminescence assay. In addition, caspase 3/7 enzymatic activity was examined after treatment. Mito-CP and 2-DG induced synergistic cytotoxicity in HepG2 cells in a dose-dependent and time-dependent manner, whereas primary cells remained viable and unaffected after treatment. The intracellular ATP levels of HepG2 cells were suppressed within 6 h of combination treatment, whereas primary cells maintained higher levels of ATP. Dose-dependent increases in caspase 3/7 activity occurred in HepG2 cells in a time-dependent manner, showing the initiation of cell death through the apoptotic pathway. These findings indicate that a combination of Mito-CP and 2-DG effectively inhibits HCC growth in vitro. The increase in caspase 3/7 activity supports the occurrence of 2-DG-induced and Mito-CP-induced apoptotic death in HCC. The inability of the compounds to induce cytotoxicity or suppress the production of ATP in primary hepatocytes provides a selective and synergistic approach for the treatment of HCC.
Collapse
|
30
|
Spielmann T, Xu L, Gad AKB, Johansson S, Widengren J. Transient state microscopy probes patterns of altered oxygen consumption in cancer cells. FEBS J 2014; 281:1317-1332. [PMID: 24418170 DOI: 10.1111/febs.12709] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/06/2013] [Accepted: 01/01/2014] [Indexed: 11/30/2022]
Abstract
Altered cellular metabolism plays an important role in many diseases, not least in many forms of cancer, where cellular metabolic pathways requiring lower oxygen consumption are often favored (the so-called Warburg effect). In this work, we have applied fluorescence-based transient state imaging and have exploited the environment sensitivity of long-lived dark states of fluorophores, in particular triplet state decay rates, to image the oxygen consumption of living cells. Our measurements can resolve differences in oxygen concentrations between different regions of individual cells, between different cell types, and also based on what metabolic pathways the cells use. In MCF-7 breast cancer cells, higher oxygen consumption can be detected when they rely on glutamine instead of glucose as their main metabolite, predominantly undergoing oxidative phosphorylation rather than glycolysis. By use of the high triplet yield dye Eosin Y the irradiance requirements during the measurements can be kept low. This reduces the instrumentation requirements, and harmful biological effects from high excitation doses can be avoided. Taken together, our imaging approach is widely applicable and capable of detecting subtle changes in oxygen consumption in live cells, stemming from the Warburg effect or reflecting other differences in the cellular metabolism. This may lead to new diagnostic means as well as advance our understanding of the interplay between cellular metabolism and major disease categories, such as cancer.
Collapse
Affiliation(s)
- Thiemo Spielmann
- Experimental Biomolecular Physics, Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
31
|
Seyfried TN, Flores RE, Poff AM, D'Agostino DP. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis 2013; 35:515-27. [PMID: 24343361 PMCID: PMC3941741 DOI: 10.1093/carcin/bgt480] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Emerging evidence indicates that cancer is primarily a metabolic disease involving disturbances in energy production through respiration and fermentation. The genomic instability observed in tumor cells and all other recognized hallmarks of cancer are considered downstream epiphenomena of the initial disturbance of cellular energy metabolism. The disturbances in tumor cell energy metabolism can be linked to abnormalities in the structure and function of the mitochondria. When viewed as a mitochondrial metabolic disease, the evolutionary theory of Lamarck can better explain cancer progression than can the evolutionary theory of Darwin. Cancer growth and progression can be managed following a whole body transition from fermentable metabolites, primarily glucose and glutamine, to respiratory metabolites, primarily ketone bodies. As each individual is a unique metabolic entity, personalization of metabolic therapy as a broad-based cancer treatment strategy will require fine-tuning to match the therapy to an individual’s unique physiology.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA and
| | | | | | | |
Collapse
|
32
|
Sullivan K, El-Hoss J, Quinlan KGR, Deo N, Garton F, Seto JTC, Gdalevitch M, Turner N, Cooney GJ, Kolanczyk M, North KN, Little DG, Schindeler A. NF1 is a critical regulator of muscle development and metabolism. Hum Mol Genet 2013; 23:1250-9. [PMID: 24163128 DOI: 10.1093/hmg/ddt515] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is emerging evidence for reduced muscle function in children with neurofibromatosis type 1 (NF1). We have examined three murine models featuring NF1 deficiency in muscle to study the effect on muscle function as well as any underlying pathophysiology. The Nf1(+/-) mouse exhibited no differences in overall weight, lean tissue mass, fiber size, muscle weakness as measured by grip strength or muscle atrophy-recovery with limb disuse, although this model lacks many other characteristic features of the human disease. Next, muscle-specific knockout mice (Nf1muscle(-/-)) were generated and they exhibited a failure to thrive leading to neonatal lethality. Intramyocellular lipid accumulations were observed by electron microscopy and Oil Red O staining. More mature muscle specimens lacking Nf1 expression taken from the limb-specific Nf1Prx1(-/-) conditional knockout line showed a 10-fold increase in muscle triglyceride content. Enzyme assays revealed a significant increase in the activities of oxidative metabolism enzymes in the Nf1Prx1(-/-) mice. Western analyses showed increases in the expression of fatty acid synthase and the hormone leptin, as well as decreased expression of a number of fatty acid transporters in this mouse line. These data support the hypothesis that NF1 is essential for normal muscle function and survival and are the first to suggest a direct link between NF1 and mitochondrial fatty acid metabolism.
Collapse
|
33
|
Sánchez-Aragó M, Formentini L, Cuezva JM. Mitochondria-mediated energy adaption in cancer: the H(+)-ATP synthase-geared switch of metabolism in human tumors. Antioxid Redox Signal 2013; 19:285-98. [PMID: 22901241 PMCID: PMC3691914 DOI: 10.1089/ars.2012.4883] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Since the signing of the National Cancer Act in 1971, cancer still remains a major cause of death despite significant progresses made in understanding the biology and treatment of the disease. After many years of ostracism, the peculiar energy metabolism of tumors has been recognized as an additional phenotypic trait of the cancer cell. RECENT ADVANCES While the enhanced aerobic glycolysis of carcinomas has already been translated to bedside for precise tumor imaging and staging of cancer patients, accepting that an impaired bioenergetic function of mitochondria is pivotal to understand energy metabolism of tumors and in its progression is debated. However, mitochondrial bioenergetics and cell death are tightly connected. CRITICAL ISSUES Recent clinical findings indicate that H(+)-ATP synthase, a core component of mitochondrial oxidative phosphorylation, is repressed at both the protein and activity levels in human carcinomas. This review summarizes the relevance that mitochondrial function has to understand energy metabolism of tumors and explores the connection between the bioenergetic function of the organelle and the activity of mitochondria as tumor suppressors. FUTURE DIRECTIONS The reversible nature of energy metabolism in tumors highlights the relevance that the microenvironment has for tumor progression. Moreover, the stimulation of mitochondrial activity or the inhibition of glycolysis suppresses tumor growth. Future research should elucidate the mechanisms promoting the silencing of oxidative phosphorylation in carcinomas. The aim is the development of new therapeutic strategies tackling energy metabolism to eradicate tumors or at least, to maintain tumor dormancy and transform cancer into a chronic disease.
Collapse
Affiliation(s)
- María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Centro de Investigación Biomédica en Red de Enfermedades Raras, Centro de Investigación Hospital 12 de Octubre, Madrid, Spain
| | | | | |
Collapse
|
34
|
van Horssen R, Willemse M, Haeger A, Attanasio F, Güneri T, Schwab A, Stock CM, Buccione R, Fransen JAM, Wieringa B. Intracellular NAD(H) levels control motility and invasion of glioma cells. Cell Mol Life Sci 2013; 70:2175-90. [PMID: 23307072 PMCID: PMC11113314 DOI: 10.1007/s00018-012-1249-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/29/2012] [Accepted: 12/17/2012] [Indexed: 12/15/2022]
Abstract
Oncogenic transformation involves reprogramming of cell metabolism, whereby steady-state levels of intracellular NAD(+) and NADH can undergo dramatic changes while ATP concentration is generally well maintained. Altered expression of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of NAD(+)-salvage, accompanies the changes in NAD(H) during tumorigenesis. Here, we show by genetic and pharmacological inhibition of NAMPT in glioma cells that fluctuation in intracellular [NAD(H)] differentially affects cell growth and morphodynamics, with motility/invasion capacity showing the highest sensitivity to [NAD(H)] decrease. Extracellular supplementation of NAD(+) or re-expression of NAMPT abolished the effects. The effects of NAD(H) decrease on cell motility appeared parallel coupled with diminished pyruvate-lactate conversion by lactate dehydrogenase (LDH) and with changes in intracellular and extracellular pH. The addition of lactic acid rescued and knockdown of LDH-A replicated the effects of [NAD(H)] on motility. Combined, our observations demonstrate that [NAD(H)] is an important metabolic component of cancer cell motility. Nutrient or drug-mediated modulation of NAD(H) levels may therefore represent a new option for blocking the invasive behavior of tumors.
Collapse
Affiliation(s)
- Remco van Horssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jose C, Melser S, Benard G, Rossignol R. Mitoplasticity: adaptation biology of the mitochondrion to the cellular redox state in physiology and carcinogenesis. Antioxid Redox Signal 2013; 18:808-49. [PMID: 22989324 DOI: 10.1089/ars.2011.4357] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adaptation and transformation biology of the mitochondrion to redox status is an emerging domain of physiology and pathophysiology. Mitochondrial adaptations occur in response to accidental changes in cellular energy demand or supply while mitochondrial transformations are a part of greater program of cell metamorphosis. The possible role of mitochondrial adaptations and transformations in pathogenesis remains unexplored, and it has become critical to decipher the stimuli and the underlying molecular pathways. Immediate activation of mitochondrial function was described during acute exercise, respiratory chain injury, Endoplasmic Reticulum stress, genotoxic stress, or environmental toxic insults. Delayed adaptations of mitochondrial form, composition, and functions were evidenced for persistent changes in redox status as observed in endurance training, in fibroblasts grown in presence of respiratory chain inhibitors or in absence of glucose, in the smooth muscle of patients with severe asthma, or in the skeletal muscle of patients with a mitochondrial disease. Besides, mitochondrial transformations were observed in the course of human cell differentiation, during immune response activation, or in cells undergoing carcinogenesis. Little is known on the signals and downstream pathways that govern mitochondrial adaptations and transformations. Few adaptative loops, including redox sensors, kinases, and transcription factors were deciphered, but their implication in physiology and pathology remains elusive. Mitoplasticity could play a protective role against aging, diabetes, cancer, or neurodegenerative diseases. Research on adaptation and transformation could allow the design of innovative therapies, notably in cancer.
Collapse
Affiliation(s)
- Caroline Jose
- University Bordeaux, Maladies Rares: Génétique et Métabolisme, France
| | | | | | | |
Collapse
|
36
|
van Horssen R, Buccione R, Willemse M, Cingir S, Wieringa B, Attanasio F. Cancer cell metabolism regulates extracellular matrix degradation by invadopodia. Eur J Cell Biol 2013; 92:113-21. [PMID: 23306026 DOI: 10.1016/j.ejcb.2012.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/20/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
Transformed cancer cells have an altered metabolism, characterized by a shift towards aerobic glycolysis, referred to as 'the Warburg phenotype'. A change in flux through mitochondrial OXPHOS and cytosolic pathways for ATP production and a gain of capacity for biomass production in order to sustain the needs for altered growth and morphodynamics are typically involved in this global rewiring of cancer cell metabolism. Characteristically, these changes in metabolism are accompanied by enhanced uptake of nutrients like glucose and glutamine. Here we focus on the relationship between cell metabolism and cell dynamics, in particular the formation and function of invadopodia, specialized structures for focal degradation of the extracellular matrix. Since we recently found presence of enzymes that are active in glycolysis and associated pathways in invadopodia, we hypothesize that metabolic adaptation and invadopodia formation are linked processes. We give an overview on the background for this idea and show for the first time that extracellular matrix degradation by invadopodia can be differentially manipulated, without effects on cell proliferation, by use of metabolic inhibitors or changes in nutrient composition of cell culture media. We conclude that cell metabolism and carbohydrate availability, especially pyruvate, are involved in fuelling of invadopodia formation and activity.
Collapse
Affiliation(s)
- Remco van Horssen
- Department of Cell Biology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
37
|
Oxidative stress and cancer: an overview. Ageing Res Rev 2013; 12:376-90. [PMID: 23123177 DOI: 10.1016/j.arr.2012.10.004] [Citation(s) in RCA: 943] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/16/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022]
Abstract
Reactive species, which mainly include reactive oxygen species (ROS), are products generated as a consequence of metabolic reactions in the mitochondria of eukaryotic cells. In normal cells, low-level concentrations of these compounds are required for signal transduction before their elimination. However, cancer cells, which exhibit an accelerated metabolism, demand high ROS concentrations to maintain their high proliferation rate. Different ways of developing ROS resistance include the execution of alternative pathways, which can avoid large amounts of ROS accumulation without compromising the energy demand required by cancer cells. Examples of these processes include the guidance of the glycolytic pathway into the pentose phosphate pathway (PPP) and/or the generation of lactate instead of employing aerobic respiration in the mitochondria. Importantly, ROS levels can be used as a thermostat to monitor the damage that cells can bear. The implications for ROS regulation are highly significant for cancer therapy because commonly used radio- and chemotherapeutic drugs influence tumor outcome through ROS modulation. Moreover, the discovery of novel biomarkers that are able to predict the clinical response to pro-oxidant therapies is a crucial challenge to overcome to allow for the personalization of cancer therapies.
Collapse
|
38
|
Goo CK, Lim HY, Ho QS, Too HP, Clement MV, Wong KP. PTEN/Akt signaling controls mitochondrial respiratory capacity through 4E-BP1. PLoS One 2012; 7:e45806. [PMID: 23049865 PMCID: PMC3458951 DOI: 10.1371/journal.pone.0045806] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 08/24/2012] [Indexed: 11/30/2022] Open
Abstract
Akt, a serine/threonine kinase has been shown to stimulate glycolysis in cancer cells but its role in mitochondrial respiration is unknown. Using PTEN-knockout mouse embryonic fibroblasts (MEFPTEN−/−) with hyper-activated Akt as a cell model, we observed a higher respiratory capacity in MEFPTEN−/− compared to the wildtype (MEFWT). The respiratory phenotype observed in MEFPTEN−/− was reproduced in MEFWT by gene silencing of PTEN which substantiated its role in regulating mitochondrial function. The increased activities of the respiratory complexes (RCs) I, III and IV were retained in the same relative proportions as those present in MEFWT, alluding to a possible co-ordinated regulation by PTEN/Akt. Using LY294002 (a PI3K inhibitor) and Akt inhibitor IV, we showed that the regulation of enzyme activities and protein expressions of the RCs was dependent on PI3K/Akt. There was insignificant difference in the protein expressions of mitochondrial transcription factor: peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and its downstream targets, the nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (mtTFA) between MEFPTEN−/− and MEFWT. Similarly, mRNA levels of the same subunits of the RCs detected in Western blots were not significantly different between MEFPTEN−/− and MEFWT suggesting that the regulation by Akt on mitochondrial function was probably not via gene transcription. On the other hand, a decrease of total 4E-BP1 with a higher expression of its phosphorylated form relative to total 4E-BP1 was found in MEFPTEN−/−, which inferred that the regulation of mitochondrial respiratory activities by Akt was in part through this protein translation pathway. Notably, gene silencing of 4E-BP1 up-regulated the protein expressions of all RCs and the action of 4E-BP1 appeared to be specific to these mitochondrial proteins. In conclusion, PTEN inactivation bestowed a bioenergetic advantage to the cells by up-regulating mitochondrial respiratory capacity through the 4E-BP1-mediated protein translation pathway.
Collapse
Affiliation(s)
- Chong Kiat Goo
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
| | - Hwee Ying Lim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
| | - Qin Shi Ho
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
| | - Heng-Phon Too
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
| | - Marie-Veronique Clement
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Kent Ridge, Singapore, Singapore
| | - Kim Ping Wong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore, Singapore
- * E-mail:
| |
Collapse
|
39
|
Alberghina L, Gaglio D, Gelfi C, Moresco RM, Mauri G, Bertolazzi P, Messa C, Gilardi MC, Chiaradonna F, Vanoni M. Cancer cell growth and survival as a system-level property sustained by enhanced glycolysis and mitochondrial metabolic remodeling. Front Physiol 2012; 3:362. [PMID: 22988443 PMCID: PMC3440026 DOI: 10.3389/fphys.2012.00362] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 08/23/2012] [Indexed: 12/14/2022] Open
Abstract
Systems Biology holds that complex cellular functions are generated as system-level properties endowed with robustness, each involving large networks of molecular determinants, generally identified by “omics” analyses. In this paper we describe four basic cancer cell properties that can easily be investigated in vitro: enhanced proliferation, evasion from apoptosis, genomic instability, and inability to undergo oncogene-induced senescence. Focusing our analysis on a K-ras dependent transformation system, we show that enhanced proliferation and evasion from apoptosis are closely linked, and present findings that indicate how a large metabolic remodeling sustains the enhanced growth ability. Network analysis of transcriptional profiling gives the first indication on this remodeling, further supported by biochemical investigations and metabolic flux analysis (MFA). Enhanced glycolysis, down-regulation of TCA cycle, decoupling of glucose and glutamine utilization, with increased reductive carboxylation of glutamine, so to yield a sustained production of growth building blocks and glutathione, are the hallmarks of enhanced proliferation. Low glucose availability specifically induces cell death in K-ras transformed cells, while PKA activation reverts this effect, possibly through at least two mitochondrial targets. The central role of mitochondria in determining the two investigated cancer cell properties is finally discussed. Taken together the findings reported herein indicate that a system-level property is sustained by a cascade of interconnected biochemical pathways that behave differently in normal and in transformed cells.
Collapse
Affiliation(s)
- Lilia Alberghina
- SysBio Centre for Systems Biology Milano and Rome, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gerrits L, Overheul GJ, Derks RC, Wieringa B, Hendriks WJ, Wansink DG. Gene duplication and conversion events shaped three homologous, differentially expressed myosin regulatory light chain (MLC2) genes. Eur J Cell Biol 2012; 91:629-39. [DOI: 10.1016/j.ejcb.2012.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 01/30/2012] [Accepted: 02/03/2012] [Indexed: 10/28/2022] Open
|
41
|
Kaambre T, Chekulayev V, Shevchuk I, Karu-Varikmaa M, Timohhina N, Tepp K, Bogovskaja J, Kütner R, Valvere V, Saks V. Metabolic control analysis of cellular respiration in situ in intraoperational samples of human breast cancer. J Bioenerg Biomembr 2012; 44:539-58. [PMID: 22836527 DOI: 10.1007/s10863-012-9457-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/21/2012] [Indexed: 12/19/2022]
Abstract
The aim of this study was to analyze quantitatively cellular respiration in intraoperational tissue samples taken from human breast cancer (BC) patients. We used oxygraphy and the permeabilized cell techniques in combination with Metabolic Control Analysis (MCA) to measure a corresponding flux control coefficient (FCC). The activity of all components of ATP synthasome, and respiratory chain complexes was found to be significantly increased in human BC cells in situ as compared to the adjacent control tissue. FCC(s) were determined upon direct activation of respiration with exogenously-added ADP and by titrating the complexes with their specific inhibitors to stepwise decrease their activity. MCA showed very high sensitivity of all complexes and carriers studied in human BC cells to inhibition as compared to mitochondria in normal oxidative tissues. The sum of FCC(s) for all ATP synthasome and respiratory chain components was found to be around 4, and the value exceeded significantly that for normal tissue (close to 1). In BC cells, the key sites of the regulation of respiration are Complex IV (FCC = 0.74), ATP synthase (FCC = 0.61), and phosphate carrier (FCC = 0.60); these FCC(s) exceed considerably (~10-fold) those for normal oxidative tissues. In human BC cells, the outer mitochondrial membrane is characterized by an increased permeability towards adenine nucleotides, the mean value of the apparent K(m) for ADP being equal to 114.8 ± 13.6 μM. Our data support the two-compartment hypothesis of tumor metabolism, the high sum of FCC(s) showing structural and functional organization of mitochondrial respiratory chain and ATP synthasome as supercomplexes in human BC.
Collapse
Affiliation(s)
- Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Estonia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Noch E, Khalili K. Oncogenic viruses and tumor glucose metabolism: like kids in a candy store. Mol Cancer Ther 2012; 11:14-23. [PMID: 22234809 DOI: 10.1158/1535-7163.mct-11-0517] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Oncogenic viruses represent a significant public health burden in light of the multitude of malignancies that result from chronic or spontaneous viral infection and transformation. Although many of the molecular signaling pathways that underlie virus-mediated cellular transformation are known, the impact of these viruses on metabolic signaling and phenotype within proliferating tumor cells is less well understood. Whether the interaction of oncogenic viruses with metabolic signaling pathways involves enhanced glucose uptake and glycolysis (both hallmark features of transformed cells) or dysregulation of molecular pathways that regulate oxidative stress, viruses are adept at facilitating tumor expansion. Through their effects on cell proliferation pathways, such as the PI3K and MAPK pathways, the cell cycle regulatory proteins p53 and ATM, and the cell stress response proteins HIF-1α and AMPK, viruses exert control over critical metabolic signaling cascades. Additionally, oncogenic viruses modulate the tumor metabolomic profile through direct and indirect interactions with glucose transporters, such as GLUT1, and specific glycolytic enzymes, including pyruvate kinase, glucose 6-phosphate dehydrogenase, and hexokinase. Through these pathways, oncogenic viruses alter the phenotypic characteristics and energy-use methods of transformed cells; therefore, it may be possible to develop novel antiglycolytic therapies to target these dysregulated pathways in virus-derived malignancies.
Collapse
Affiliation(s)
- Evan Noch
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | |
Collapse
|
43
|
Ma X, Sim SJ. Ultrasensitive detection of the reduced form of nicotinamide adenine dinucleotide based on carbon nanotube field effect transistor. Analyst 2012; 137:3328-34. [PMID: 22669083 DOI: 10.1039/c2an16253a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We developed a simple, ultrasensitive, and quantitative detection method for the reduced form of nicotinamide adenine dinucleotide (NADH), based on carbon nanotube field effect transistors (CNTFETs). Following the injection of NADH at different concentrations, we obtained different electrical signals from a semiconductor characterization system mimicking biological catalysis of NADH dehydrogenase (CoI). Here, FET was fabricated via photolithography, attaching silicon wells, as the detection chamber, on the channel area of the single wall carbon nanotube (SWCNT). SWCNTs were functionalized with phenazine derivant, a counterpart of the key functional prosthetic group of CoI enzyme. In the presence of NADH, electrons transferred to phenazine derivant through SWCNT, by analogous means of the electron transport chain formed by a series of iron-sulfur (FeS) clusters in CoI. Using this method, the limit of detection was as low as 1 pM, and the range of linear response was 10 pM to 500 nM. Significantly, this approach possesses great potential for applications in real-time detection of NADH at extremely low concentrations, and rigorous analysis for NADH in electrochemical fields.
Collapse
Affiliation(s)
- Xingyi Ma
- Department of Chemical and Biological Engineering, Korea University, Seoul, 136-713, Republic of Korea
| | | |
Collapse
|
44
|
Measuring energy metabolism in cultured cells, including human pluripotent stem cells and differentiated cells. Nat Protoc 2012; 7:1068-85. [PMID: 22576106 DOI: 10.1038/nprot.2012.048] [Citation(s) in RCA: 216] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Measurements of glycolysis and mitochondrial function are required to quantify energy metabolism in a wide variety of cellular contexts. In human pluripotent stem cells (hPSCs) and their differentiated progeny, this analysis can be challenging because of the unique cell properties, growth conditions and expense required to maintain these cell types. Here we provide protocols for analyzing energy metabolism in hPSCs and their early differentiated progenies that are generally applicable to mature cell types as well. Our approach has revealed distinct energy metabolism profiles used by hPSCs, differentiated cells, a variety of cancer cells and Rho-null cells. The protocols measure or estimate glycolysis on the basis of the extracellular acidification rate, and they measure or estimate oxidative phosphorylation on the basis of the oxygen consumption rate. Assays typically require 3 h after overnight sample preparation. Companion methods are also discussed and provided to aid researchers in developing more sophisticated experimental regimens for extended analyses of cellular bioenergetics.
Collapse
|
45
|
Cheng G, Zielonka J, Dranka BP, McAllister D, Mackinnon AC, Joseph J, Kalyanaraman B. Mitochondria-targeted drugs synergize with 2-deoxyglucose to trigger breast cancer cell death. Cancer Res 2012; 72:2634-44. [PMID: 22431711 DOI: 10.1158/0008-5472.can-11-3928] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cancer cells are long known to exhibit increased aerobic glycolysis, but glycolytic inhibition has not offered a viable chemotherapeutic strategy in part because of the systemic toxicity of antiglycolytic agents. However, recent studies suggest that a combined inhibition of glycolysis and mitochondrial function may help overcome this issue. In this study, we investigated the chemotherapeutic efficacies of mitochondria-targeted drugs (MTD) in combination with 2-deoxy-d-glucose (2-DG), a compound that inhibits glycolysis. Using the MTDs, termed Mito-CP and Mito-Q, we evaluated relative cytotoxic effects and mitochondrial bioenergetic changes in vitro. Interestingly, both Mito-CP and Mito-Q synergized with 2-DG to decrease ATP levels in two cell lines. However, with time, the cellular bioenergetic function and clonogenic survival were largely restored in some cells. In a xenograft model of human breast cancer, combined treatment of Mito-CP and 2-DG led to significant tumor regression in the absence of significant morphologic changes in kidney, liver, or heart. Collectively, our findings suggest that dual targeting of mitochondrial bioenergetic metabolism with MTDs and glycolytic inhibitors such as 2-DG may offer a promising chemotherapeutic strategy.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Sotgia F, Martinez-Outschoorn UE, Howell A, Pestell RG, Pavlides S, Lisanti MP. Caveolin-1 and cancer metabolism in the tumor microenvironment: markers, models, and mechanisms. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2011; 7:423-67. [PMID: 22077552 DOI: 10.1146/annurev-pathol-011811-120856] [Citation(s) in RCA: 229] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Caveolins are a family of membrane-bound scaffolding proteins that compartmentalize and negatively regulate signal transduction. Recent studies have implicated a loss of caveolin-1 (Cav-1) expression in the pathogenesis of human cancers. Loss of Cav-1 expression in cancer-associated fibroblasts results in an activated tumor microenvironment, thereby driving early tumor recurrence, metastasis, and poor clinical outcome in breast and prostate cancers. We describe various paracrine signaling mechanism(s) by which the loss of stromal Cav-1 promotes tumor progression, including fibrosis, extracellular matrix remodeling, and the metabolic/catabolic reprogramming of cancer-associated fibroblast, to fuel the growth of adjacent tumor cells. It appears that oxidative stress is the root cause of initiation of the loss of stromal Cav-1 via autophagy, which provides further impetus for the use of antioxidants in anticancer therapy. Finally, we discuss the functional role of Cav-1 in epithelial cancer cells.
Collapse
Affiliation(s)
- Federica Sotgia
- The Jefferson Stem Cell Biology and Regenerative Medicine Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | | | | | | | |
Collapse
|
47
|
The awakening of an advanced malignant cancer: an insult to the mitochondrial genome. Biochim Biophys Acta Gen Subj 2011; 1820:652-62. [PMID: 21920409 DOI: 10.1016/j.bbagen.2011.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 08/26/2011] [Accepted: 08/29/2011] [Indexed: 02/08/2023]
Abstract
BACKGROUND In only months-to-years a primary cancer can progress to an advanced phenotype that is metastatic and resistant to clinical treatments. As early as the 1900s, it was discovered that the progression of a cancer to the advanced phenotype is often associated with a shift in the metabolic profile of the disease from a state of respiration to anaerobic fermentation - a phenomenon denoted as the Warburg Effect. SCOPE OF REVIEW Reports in the literature strongly suggest that the Warburg Effect is generated as a response to a loss in the integrity of the sequence and/or copy number of the mitochondrial genome content within a cancer. MAJOR CONCLUSIONS Multiple studies regarding the progression of cancer indicate that mutation, and/or, a flux in the copy number, of the mitochondrial genome content can support the early development of a cancer, until; the mutational load and/or the reduction-to-depletion of the copy number of the mitochondrial genome content induces the progression of the disease to an advanced phenotype. GENERAL SIGNIFICANCE Collectively, evidence has revealed that the human cell has incorporated the mitochondrial genome content into a cellular mechanism that, when pathologically actuated, can de(un)differentiate a cancer from the parental tissue of origin into an autonomous disease that disrupts the hierarchical structure-and-function of the human body. This article is part of a Special Issue entitled: Biochemistry of Mitochondria.
Collapse
|
48
|
Wagner BA, Venkataraman S, Buettner GR. The rate of oxygen utilization by cells. Free Radic Biol Med 2011; 51:700-12. [PMID: 21664270 PMCID: PMC3147247 DOI: 10.1016/j.freeradbiomed.2011.05.024] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 04/26/2011] [Accepted: 05/19/2011] [Indexed: 11/18/2022]
Abstract
The discovery of oxygen is considered by some to be the most important scientific discovery of all time--from both physical-chemical/astrophysics and biology/evolution viewpoints. One of the major developments during evolution is the ability to capture dioxygen in the environment and deliver it to each cell in the multicellular, complex mammalian body-on demand, i.e., just in time. Humans use oxygen to extract approximately 2550 calories (10.4 MJ) from food to meet daily energy requirements. This combustion requires about 22 mol of dioxygen per day, or 2.5×10(-4) mol s(-1). This is an average rate of oxygen utilization of 2.5×10(-18) mol cell(-1) s(-1), i.e., 2.5 amol cell(-1) s(-1). Cells have a wide range of oxygen utilization, depending on cell type, function, and biological status. Measured rates of oxygen utilization by mammalian cells in culture range from <1 to >350 amol cell(-1) s(-1). There is a loose positive linear correlation of the rate of oxygen consumption by mammalian cells in culture with cell volume and cell protein. The use of oxygen by cells and tissues is an essential aspect of the basic redox biology of cells and tissues. This type of quantitative information is fundamental to investigations in quantitative redox biology, especially redox systems biology.
Collapse
Affiliation(s)
- Brett A Wagner
- Free Radical and Radiation Biology Program and ESR Facility, The University of Iowa, Iowa City, IA 52242–1181, USA
| | | | | |
Collapse
|
49
|
Jose C, Bellance N, Rossignol R. Choosing between glycolysis and oxidative phosphorylation: A tumor's dilemma? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2011; 1807:552-61. [DOI: 10.1016/j.bbabio.2010.10.012] [Citation(s) in RCA: 340] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 10/11/2010] [Accepted: 10/11/2010] [Indexed: 12/25/2022]
|
50
|
Fisher KW, Das B, Kortum RL, Chaika OV, Lewis RE. Kinase suppressor of ras 1 (KSR1) regulates PGC1α and estrogen-related receptor α to promote oncogenic Ras-dependent anchorage-independent growth. Mol Cell Biol 2011; 31:2453-61. [PMID: 21518958 PMCID: PMC3133429 DOI: 10.1128/mcb.05255-11] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 04/08/2011] [Indexed: 12/20/2022] Open
Abstract
Kinase suppressor of ras 1 (KSR1) is a molecular scaffold of the Raf/MEK/extracellular signal-regulated kinase (ERK) cascade that enhances oncogenic Ras signaling. Here we show KSR1-dependent, but ERK-independent, regulation of metabolic capacity is mediated through the expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) and estrogen-related receptor α (ERRα). This KSR1-regulated pathway is essential for the transformation of cells by oncogenic Ras. In mouse embryo fibroblasts (MEFs) expressing H-Ras(V12), ectopic PGC1α was sufficient to rescue ERRα expression, metabolic capacity, and anchorage-independent growth in the absence of KSR1. The ability of PGC1α to promote anchorage-independent growth required interaction with ERRα, and treatment with an inhibitor of ERRα impeded anchorage-independent growth. In contrast to PGC1α, the expression of constitutively active ERRα (CA-ERRα) was sufficient to enhance metabolic capacity but not anchorage-independent growth in the absence of KSR1. These data reveal KSR1-dependent control of PGC1α- and ERRα-dependent pathways that are necessary and sufficient for signaling by oncogenic H-Ras(V12) to regulate metabolism and anchorage-independent growth, providing novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Kurt W. Fisher
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Binita Das
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | | | - Oleg V. Chaika
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Robert E. Lewis
- Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|