1
|
Souza PFN, Zelaya EAE, da Silva EL, Brasil-Oliveira LL, de Oliveira FL, de Moraes MEA, Montenegro RC, Mesquita FP. PepGAT, a chitinase-derived peptide, alters the proteomic profile of colorectal cancer cells and perturbs pathways involved in cancer survival. Int J Biol Macromol 2025; 299:140204. [PMID: 39848367 DOI: 10.1016/j.ijbiomac.2025.140204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Colorectal cancer (CRC) affects the population worldwide, occupying the first place in terms of death and incidence. Synthetic peptides (SPs) emerged as alternative molecules due to their activity and low toxicity. Proteomic analysis of PepGAT-treated HCT-116 cells revealed a decreased abundance of proteins involved in ROS metabolism and energetic metabolisms, cell cycle, DNA repair, migration, invasion, cancer aggressiveness, and proteins involved in resistance to 5-FU. PepGAT induced earlier ROS and apoptosis in HCT-116 cells, cell cycle arrest, and inhibited HCT-116 migration. PepGAT enhances the action of 5-FU against HCT-116 cells by dropping down 6-fold the 5-FU toward HCT-116 and reduces its toxicity for non-cancerous cells. These findings strongly suggest the multiple mechanisms of action displayed by PepGAT against CRC cells and its potential to either be studied alone or in combination with 5-FU to develop new studies against CRC and might develop new drugs against it.
Collapse
Affiliation(s)
- Pedro Filho Noronha Souza
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Cearense Foundation to Support Scientific and Technological Development, Brazil.
| | - Elmer Adilson Espino Zelaya
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Emerson Lucena da Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Laís Lacerda Brasil-Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Francisco Laio de Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil.
| |
Collapse
|
2
|
Raju C, Sankaranarayanan K. Insights on post-translational modifications in fatty liver and fibrosis progression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167659. [PMID: 39788217 DOI: 10.1016/j.bbadis.2025.167659] [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: 09/06/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease [MASLD] is a pervasive multifactorial health burden. Post-translational modifications [PTMs] of amino acid residues in protein domains demonstrate pivotal roles for imparting dynamic alterations in the cellular micro milieu. The crux of identifying novel druggable targets relies on comprehensively studying the etiology of metabolic disorders. This review article presents how different chemical moieties of various PTMs like phosphorylation, methylation, ubiquitination, glutathionylation, neddylation, acetylation, SUMOylation, lactylation, crotonylation, hydroxylation, glycosylation, citrullination, S-sulfhydration and succinylation presents the cause-effect contribution towards the MASLD spectra. Additionally, the therapeutic prospects in the management of liver steatosis and hepatic fibrosis via targeting PTMs and regulatory enzymes are also encapsulated. This review seeks to understand the function of protein modifications in progression and promote the markers discovery of diagnostic, prognostic and drug targets towards MASLD management which could also halt the progression of a catalogue of related diseases.
Collapse
Affiliation(s)
- Chithra Raju
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India.
| |
Collapse
|
3
|
Rahman AFMT, Benko A, Bulbule S, Gottschalk CG, Arnold LA, Roy A. Tetrahydrobiopterin in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Friend or Foe? Biomolecules 2025; 15:102. [PMID: 39858496 PMCID: PMC11763651 DOI: 10.3390/biom15010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Myalgic Encephalomyelitis or Chronic Fatigue Syndrome (ME/CFS) is a chronic multisystem disease characterized by severe muscle fatigue, pain, dizziness, and brain fog. The two most common symptoms are post-exertional malaise (PEM) and orthostatic intolerance (OI). ME/CFS patients with OI (ME+OI) suffer from dizziness or faintness due to a sudden drop in blood pressure while maintaining an upright posture. Clinical research has demonstrated that patients with OI display severe cardiovascular abnormalities resulting in reduced effective blood flow in the cerebral blood vessels. However, despite intense investigation, it is not known why the effective cerebral blood flow is reduced in OI patients. Based on our recent findings, we observed that tetrahydrobiopterin (BH4) metabolism was highly dysregulated in ME+OI patients. In the current review article, we attempted to summarize our recent findings on BH4 metabolism to shed light on the molecular mechanisms of OI.
Collapse
Affiliation(s)
- A. F. M. Towheedur Rahman
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Anna Benko
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Sarojini Bulbule
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
| | - Carl Gunnar Gottschalk
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - Leggy A. Arnold
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
| | - Avik Roy
- Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA; (A.F.M.T.R.); (A.B.); (L.A.A.)
- Research and Development Laboratory, Chemistry Building, 2000 E Kenwood Blvd, Suite #320, Milwaukee, WI 53211, USA; (S.B.); (C.G.G.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| |
Collapse
|
4
|
Meeson KE, Schwartz JM. Constraint-based modelling predicts metabolic signatures of low and high-grade serous ovarian cancer. NPJ Syst Biol Appl 2024; 10:96. [PMID: 39181893 PMCID: PMC11344801 DOI: 10.1038/s41540-024-00418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Ovarian cancer is an aggressive, heterogeneous disease, burdened with late diagnosis and resistance to chemotherapy. Clinical features of ovarian cancer could be explained by investigating its metabolism, and how the regulation of specific pathways links to individual phenotypes. Ovarian cancer is of particular interest for metabolic research due to its heterogeneous nature, with five distinct subtypes having been identified, each of which may display a unique metabolic signature. To elucidate metabolic differences, constraint-based modelling (CBM) represents a powerful technology, inviting the integration of 'omics' data, such as transcriptomics. However, many CBM methods have not prioritised accurate growth rate predictions, and there are very few ovarian cancer genome-scale studies. Here, a novel method for CBM has been developed, employing the genome-scale model Human1 and flux balance analysis, enabling the integration of in vitro growth rates, transcriptomics data and media conditions to predict the metabolic behaviour of cells. Using low- and high-grade ovarian cancer, subtype-specific metabolic differences have been predicted, which have been supported by publicly available CRISPR-Cas9 data from the Cancer Cell Line Encyclopaedia and an extensive literature review. Metabolic drivers of aggressive, invasive phenotypes, as well as pathways responsible for increased chemoresistance in low-grade cell lines have been suggested. Experimental gene dependency data has been used to validate areas of the pentose phosphate pathway as essential for low-grade cellular growth, highlighting potential vulnerabilities for this ovarian cancer subtype.
Collapse
Affiliation(s)
- Kate E Meeson
- School of Biological Sciences, University of Manchester, Manchester, UK
| | - Jean-Marc Schwartz
- School of Biological Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
5
|
Koltai T, Fliegel L. Dichloroacetate for Cancer Treatment: Some Facts and Many Doubts. Pharmaceuticals (Basel) 2024; 17:744. [PMID: 38931411 PMCID: PMC11206832 DOI: 10.3390/ph17060744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Rarely has a chemical elicited as much controversy as dichloroacetate (DCA). DCA was initially considered a dangerous toxic industrial waste product, then a potential treatment for lactic acidosis. However, the main controversies started in 2008 when DCA was found to have anti-cancer effects on experimental animals. These publications showed contradictory results in vivo and in vitro such that a thorough consideration of this compound's in cancer is merited. Despite 50 years of experimentation, DCA's future in therapeutics is uncertain. Without adequate clinical trials and health authorities' approval, DCA has been introduced in off-label cancer treatments in alternative medicine clinics in Canada, Germany, and other European countries. The lack of well-planned clinical trials and its use by people without medical training has discouraged consideration by the scientific community. There are few thorough clinical studies of DCA, and many publications are individual case reports. Case reports of DCA's benefits against cancer have been increasing recently. Furthermore, it has been shown that DCA synergizes with conventional treatments and other repurposable drugs. Beyond the classic DCA target, pyruvate dehydrogenase kinase, new target molecules have also been recently discovered. These findings have renewed interest in DCA. This paper explores whether existing evidence justifies further research on DCA for cancer treatment and it explores the role DCA may play in it.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina
| | - Larry Fliegel
- Department of Biochemistry, University Alberta, Edmonton, AB T6G 2H7, Canada;
| |
Collapse
|
6
|
Gholamzad A, Khakpour N, Khosroshahi EM, Asadi S, Koohpar ZK, Matinahmadi A, Jebali A, Rashidi M, Hashemi M, Sadi FH, Gholamzad M. Cancer stem cells: The important role of CD markers, Signaling pathways, and MicroRNAs. Pathol Res Pract 2024; 256:155227. [PMID: 38490099 DOI: 10.1016/j.prp.2024.155227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/17/2024]
Abstract
For the first time, a subset of small cancer cells identified in acute myeloid leukemia has been termed Cancer Stem Cells (CSCs). These cells are notorious for their robust proliferation, self-renewal abilities, significant tumor-forming potential, spread, and resistance to treatments. CSCs are a global concern, as it found in numerous types of cancer, posing a real-world challenge today. Our review encompasses research on key CSC markers, signaling pathways, and MicroRNA in three types of cancer: breast, colon, and liver. These factors play a critical role in either promoting or inhibiting cancer cell growth. The reviewed studies have shown that as cells undergo malignant transformation, there can be an increase or decrease in the expression of different Cluster of Differentiation (CD) markers on their surface. Furthermore, alterations in essential signaling pathways, such as Wnt and Notch1, may impact CSC proliferation, survival, and movement, while also providing potential targets for cancer therapies. Additionally, some research has focused on MicroRNAs due to their dual role as potential therapeutic biomarkers and their ability to enhance CSCs' response to anti-cancer drugs. MicroRNAs also regulate a wide array of cellular processes, including the self-renewal and pluripotency of CSCs, and influence gene transcription. Thus, these studies indicate that MicroRNAs play a significant role in the malignancy of various tumors. Although the gathered information suggests that specific CSC markers, signaling pathways, and MicroRNAs are influential in determining the destiny of cancer cells and could be advantageous for therapeutic strategies, their precise roles and impacts remain incompletely defined, necessitating further investigation.
Collapse
Affiliation(s)
- Amir Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Niloofar Khakpour
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch,Islamic Azad University, Tonekabon, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus,Torun,Poland
| | - Ali Jebali
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Deprtment of Medical Nanotechnology,Faculty of Advanced Sciences and Technology,Tehran Medical Sciences,Islamic Azad University, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | | | - Mehrdad Gholamzad
- Department of Microbiology and Immunology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
8
|
Coyle JP, Johnson C, Jensen J, Farcas M, Derk R, Stueckle TA, Kornberg TG, Rojanasakul Y, Rojanasakul LW. Variation in pentose phosphate pathway-associated metabolism dictates cytotoxicity outcomes determined by tetrazolium reduction assays. Sci Rep 2023; 13:8220. [PMID: 37217524 DOI: 10.1038/s41598-023-35310-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023] Open
Abstract
Tetrazolium reduction and resazurin assays are the mainstay of routine in vitro toxicity batteries. However, potentially erroneous characterization of cytotoxicity and cell proliferation can arise if verification of baseline interaction of test article with method employed is neglected. The current investigation aimed to demonstrate how interpretation of results from several standard cytotoxicity and proliferation assays vary in dependence on contributions from the pentose phosphate pathway (PPP). Non-tumorigenic Beas-2B cells were treated with graded concentrations of benzo[a]pyrene (B[a]P) for 24 and 48 h prior to cytotoxicity and proliferation assessment with commonly used MTT, MTS, WST1, and Alamar Blue assays. B[a]P caused enhanced metabolism of each dye assessed despite reductions in mitochondrial membrane potential and was reversed by 6-aminonicotinamide (6AN)-a glucose-6-phosphate dehydrogenase inhibitor. These results demonstrate differential sensitivity of standard cytotoxicity assessments on the PPP, thus (1) decoupling "mitochondrial activity" as an interpretation of cellular formazan and Alamar Blue metabolism, and (2) demonstrating the implicit requirement for investigators to sufficiently verify interaction of these methods in routine cytotoxicity and proliferation characterization. The nuances of method-specific extramitochondrial metabolism must be scrutinized to properly qualify specific endpoints employed, particularly under the circumstances of metabolic reprogramming.
Collapse
Affiliation(s)
- Jayme P Coyle
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA.
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, 1095 Willowdale Rd., Morgantown, WV, 26505, USA.
| | - Caroline Johnson
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Jake Jensen
- Department of Environmental Health, Harvard University, Boston, MA, USA
| | - Mariana Farcas
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Raymond Derk
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Todd A Stueckle
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Tiffany G Kornberg
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA
| | - Liying W Rojanasakul
- HELD/ACIB, National Institute for Occupational Safety and Health, Morgantown, WV, USA.
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, 1095 Willowdale Rd., Morgantown, WV, 26505, USA.
| |
Collapse
|
9
|
Zaied AA, Ushio‐Fukai M, Fukai T, Kovacs‐Kasa A, Alhusban S, Sudhahar V, Ganta VC, Annex BH. Pentose Pathway Activation Is Superior to Increased Glycolysis for Therapeutic Angiogenesis in Peripheral Arterial Disease. J Am Heart Assoc 2023; 12:e027986. [PMID: 36974760 PMCID: PMC10122893 DOI: 10.1161/jaha.122.027986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 03/29/2023]
Abstract
Background In endothelial cells (ECs), glycolysis, regulated by PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, isoform-3), is the major metabolic pathway for ATP generation. In preclinical peripheral artery disease models, VEGF165a (vascular endothelial growth factor165a) and microRNA-93 both promote angiogenesis. Methods and Results Mice following hind-limb ischemia (HLI) and ECs with, and without, hypoxia and serum starvation were examined with, and without, microRNA-93 and VEGF165a. Post-HLI perfusion recovery was monitored. EC metabolism was studied using seahorse assay, and the expression and activity of major metabolism genes were assessed. Reactive oxygen species levels and EC permeability were evaluated. C57Bl/6J mice generated a robust angiogenic response to HLI, with ECs from ischemic versus nonischemic muscle demonstrating no increase in glycolysis. Balb/CJ mice generated a poor angiogenic response post-HLI; ischemic versus nonischemic ECs demonstrated significant increase in glycolysis. MicroRNA-93-treated Balb/CJ mice post-HLI showed better perfusion recovery, with ischemic versus nonischemic ECs showing no increase in glycolysis. VEGF165a-treated Balb/CJ mice post-HLI showed no improvement in perfusion recovery with ischemic versus nonischemic ECs showing significant increase in glycolysis. ECs under hypoxia and serum starvation upregulated PFKFB3. In ECs under hypoxia and serum starvation, VEGF165a versus control significantly upregulated PFKFB3 and glycolysis, whereas miR-93 versus control demonstrated no increase in PFKFB3 or glycolysis. MicroRNA-93 versus VEGF165a upregulated glucose-6-phosphate dehydrogenase expression and activity, activating the pentose phosphate pathway. MicroRNA-93 versus control increased reduced nicotinamide adenine dinucleotide phosphate and virtually eliminated the increase in reactive oxygen species. In ECs under hypoxia and serum starvation, VEGF165a significantly increased and miR-93 decreased EC permeability. Conclusions In peripheral artery disease, activation of the pentose phosphate pathway to promote angiogenesis may offer potential therapeutic advantages.
Collapse
Affiliation(s)
- Abdelrahman A. Zaied
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Masuko Ushio‐Fukai
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Tohru Fukai
- Departments of Pharmacology and ToxicologyMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Charlie Norwood Veterans Affairs Medical CenterAugustaGAUSA
| | - Anita Kovacs‐Kasa
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Suhib Alhusban
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Varadarajan Sudhahar
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Vijay C. Ganta
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Brian H. Annex
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| |
Collapse
|
10
|
Yıldız ML, Demir Y, Küfrevioğlu ÖI. Screening of in vitro and in silico effect of Fluorophenylthiourea compounds on glucose 6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase enzymes. J Mol Recognit 2022; 35:e2987. [PMID: 36326002 DOI: 10.1002/jmr.2987] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 01/05/2023]
Abstract
Inhibition studies of enzymes in the pentose phosphate pathway (PPP) have recently emerged as a promising technique for pharmacological intervention in several illnesses. Glucose 6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD) are the most important enzymes of the PPP. For this purpose, in the current study, we examined the effect of some fluorophenylthiourea on G6PD and 6PGD enzyme activity. These compounds exhibited moderate inhibitory activity against G6PD and 6PGD with KI values ranging from 21.60 ± 8.42 to 39.70 ± 11.26 μM, and 15.82 ± 1.54 to 29.97 ± 5.72 μM, respectively. 2,6-difluorophenylthiourea displayed the most potent inhibitory effect for G6PD, and 2-fluorophenylthiourea demonstrated the most substantial inhibitory effect for 6PGD. Furthermore, the molecular docking analyses of the fluorophenylthioureas, competitive inhibitors, were performed to understand the binding interactions at the enzymes' binding site.
Collapse
Affiliation(s)
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| | | |
Collapse
|
11
|
Katturajan R, Nithiyanandam S, Parthasarathy M, Valsala Gopalakrishnan A, Sathiyamoorthi E, Lee J, Ramesh T, Iyer M, Prince SE, Ganesan R. Immunomodulatory Role of Thioredoxin Interacting Protein in Cancer's Impediments: Current Understanding and Therapeutic Implications. Vaccines (Basel) 2022; 10:1902. [PMID: 36366411 PMCID: PMC9699629 DOI: 10.3390/vaccines10111902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 10/30/2023] Open
Abstract
Cancer, which killed ten million people in 2020, is expected to become the world's leading health problem and financial burden. Despite the development of effective therapeutic approaches, cancer-related deaths have increased by 25.4% in the last ten years. Current therapies promote apoptosis and oxidative stress DNA damage and inhibit inflammatory mediators and angiogenesis from providing temporary relief. Thioredoxin-binding protein (TXNIP) causes oxidative stress by inhibiting the function of the thioredoxin system. It is an important regulator of many redox-related signal transduction pathways in cells. In cancer cells, it functions as a tumor suppressor protein that inhibits cell proliferation. In addition, TXNIP levels in hemocytes increased after immune stimulation, suggesting that TXNIP plays an important role in immunity. Several studies have provided experimental evidence for the immune modulatory role of TXNIP in cancer impediments. TXNIP also has the potential to act against immune cells in cancer by mediating the JAK-STAT, MAPK, and PI3K/Akt pathways. To date, therapies targeting TXNIP in cancer are still under investigation. This review highlights the role of TXNIP in preventing cancer, as well as recent reports describing its functions in various immune cells, signaling pathways, and promoting action against cancer.
Collapse
Affiliation(s)
- Ramkumar Katturajan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Sangeetha Nithiyanandam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Manisha Parthasarathy
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | | | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Mahalaxmi Iyer
- Livestock Farming and Bioresource Technology, Coimbatore 641003, Tamil Nadu, India
| | - Sabina Evan Prince
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Raja Ganesan
- Institute for Liver and Digestive Disease, College of Medicine, Hallym University, Chuncheon 24253, Korea
| |
Collapse
|
12
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
13
|
Chen Y, Li Y. Metabolic reprogramming and immunity in cancer. CANCER IMMUNOLOGY AND IMMUNOTHERAPY 2022:137-196. [DOI: 10.1016/b978-0-12-823397-9.00006-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Harland A, Liu X, Ghirardello M, Galan MC, Perks CM, Kurian KM. Glioma Stem-Like Cells and Metabolism: Potential for Novel Therapeutic Strategies. Front Oncol 2021; 11:743814. [PMID: 34532295 PMCID: PMC8438230 DOI: 10.3389/fonc.2021.743814] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022] Open
Abstract
Glioma stem-like cells (GSCs) were first described as a population which may in part be resistant to traditional chemotherapeutic therapies and responsible for tumour regrowth. Knowledge of the underlying metabolic complexity governing GSC growth and function may point to potential differences between GSCs and the tumour bulk which could be harnessed clinically. There is an increasing interest in the direct/indirect targeting or reprogramming of GSC metabolism as a potential novel therapeutic approach in the adjuvant or recurrent setting to help overcome resistance which may be mediated by GSCs. In this review we will discuss stem-like models, interaction between metabolism and GSCs, and potential current and future strategies for overcoming GSC resistance.
Collapse
Affiliation(s)
- Abigail Harland
- Brain Tumour Research Centre, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Xia Liu
- Brain Tumour Research Centre, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Mattia Ghirardello
- Galan Research Group, School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - M Carmen Galan
- Galan Research Group, School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Claire M Perks
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Kathreena M Kurian
- Brain Tumour Research Centre, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
15
|
van Doorn CLR, Schouten GK, van Veen S, Walburg KV, Esselink JJ, Heemskerk MT, Vrieling F, Ottenhoff THM. Pyruvate Dehydrogenase Kinase Inhibitor Dichloroacetate Improves Host Control of Salmonella enterica Serovar Typhimurium Infection in Human Macrophages. Front Immunol 2021; 12:739938. [PMID: 34552598 PMCID: PMC8450447 DOI: 10.3389/fimmu.2021.739938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/23/2021] [Indexed: 01/11/2023] Open
Abstract
Global increases in the prevalence of antimicrobial resistance highlight the urgent need for novel strategies to combat infectious diseases. Recent studies suggest that host metabolic pathways play a key role in host control of intracellular bacterial pathogens. In this study we explored the potential of targeting host metabolic pathways for innovative host-directed therapy (HDT) against intracellular bacterial infections. Through gene expression profiling in human macrophages, pyruvate metabolism was identified as potential key pathway involved in Salmonella enterica serovar Typhimurium (Stm) infections. Next, the effect of targeting pyruvate dehydrogenase kinases (PDKs) - which are regulators of the metabolic checkpoint pyruvate dehydrogenase complex (PDC) - on macrophage function and bacterial control was studied. Chemical inhibition of PDKs by dichloroacetate (DCA) induced PDC activation and was accompanied with metabolic rewiring in classically activated macrophages (M1) but not in alternatively activated macrophages (M2), suggesting cell-type specific effects of dichloroacetate on host metabolism. Furthermore, DCA treatment had minor impact on cytokine and chemokine secretion on top of infection, but induced significant ROS production by M1 and M2. DCA markedly and rapidly reduced intracellular survival of Stm, but interestingly not Mycobacterium tuberculosis, in human macrophages in a host-directed manner. In conclusion, DCA represents a promising novel HDT compound targeting pyruvate metabolism for the treatment of Stm infections.
Collapse
|
16
|
Miyagaki S, Kikuchi K, Mori J, Lopaschuk GD, Iehara T, Hosoi H. Inhibition of lipid metabolism exerts antitumor effects on rhabdomyosarcoma. Cancer Med 2021; 10:6442-6455. [PMID: 34472721 PMCID: PMC8446407 DOI: 10.1002/cam4.4185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/17/2021] [Indexed: 12/14/2022] Open
Abstract
Rhabdomyosarcoma exhibits tumor-specific energy metabolic changes that include the Warburg effect. Since targeting cancer metabolism is a promising therapeutic approach, we examined the antitumor effects of suppressing lipid metabolism in rhabdomyosarcoma. We suppressed lipid metabolism in rhabdomyosarcoma cells in vitro by administering an inhibitor of malonyl-CoA decarboxylase, which increases malonyl-CoA and decreases fatty acid oxidation. Suppression of lipid metabolism in rhabdomyosarcoma cells decreased cell proliferation by inducing cell cycle arrest. Metabolomic analysis showed an increase in glycolysis and inactivation of the pentose phosphate pathway. Immunoblotting analysis revealed upregulated expression of the autophagy marker LC3A/B-II due to increased phosphorylation of AMP-activated protein kinase, a nutrient sensor. p21 protein expression level also increased. Inhibition of both lipid metabolism and autophagy suppressed tumor proliferation and increased apoptosis. In vivo studies involved injection of human Rh30 cells into the gastrocnemius muscle of 6-week-old female nude mice, which were divided into normal chow and low-fat diet groups. The mice fed a low-fat diet for 21 days showed reduced tumor growth compared to normal chow diet-fed mice. Suppression of lipid metabolism disrupted the equilibrium of the cancer-specific metabolism in rhabdomyosarcoma, resulting in a tumor growth-inhibition effect. Therefore, the development of treatments focusing on the lipid dependence of rhabdomyosarcoma is highly promising.
Collapse
Affiliation(s)
- Satoshi Miyagaki
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Kikuchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Pediatrics, Uji Takeda Hospital, Kyoto, Japan
| | - Jun Mori
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Gary D Lopaschuk
- Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Salnikova D, Orekhova V, Grechko A, Starodubova A, Bezsonov E, Popkova T, Orekhov A. Mitochondrial Dysfunction in Vascular Wall Cells and Its Role in Atherosclerosis. Int J Mol Sci 2021; 22:8990. [PMID: 34445694 PMCID: PMC8396504 DOI: 10.3390/ijms22168990] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022] Open
Abstract
Altered mitochondrial function is currently recognized as an important factor in atherosclerosis initiation and progression. Mitochondrial dysfunction can be caused by mitochondrial DNA (mtDNA) mutations, which can be inherited or spontaneously acquired in various organs and tissues, having more or less profound effects depending on the tissue energy status. Arterial wall cells are among the most vulnerable to mitochondrial dysfunction due to their barrier and metabolic functions. In atherosclerosis, mitochondria cause alteration of cellular metabolism and respiration and are known to produce excessive amounts of reactive oxygen species (ROS) resulting in oxidative stress. These processes are involved in vascular disease and chronic inflammation associated with atherosclerosis. Currently, the list of known mtDNA mutations associated with human pathologies is growing, and many of the identified mtDNA variants are being tested as disease markers. Alleviation of oxidative stress and inflammation appears to be promising for atherosclerosis treatment. In this review, we discuss the role of mitochondrial dysfunction in atherosclerosis development, focusing on the key cell types of the arterial wall involved in the pathological processes. Accumulation of mtDNA mutations in isolated arterial wall cells, such as endothelial cells, may contribute to the development of local inflammatory process that helps explaining the focal distribution of atherosclerotic plaques on the arterial wall surface. We also discuss antioxidant and anti-inflammatory approaches that can potentially reduce the impact of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Diana Salnikova
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
- Laboratory of Oncoproteomics, Institute of Carconigenesis, N. N. Blokhin Cancer Research Centre, 115478 Moscow, Russia
| | - Varvara Orekhova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (E.B.); (A.O.)
| | - Andrey Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, 109240 Moscow, Russia;
| | - Antonina Starodubova
- Federal Research Centre for Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia;
- Therapy Faculty, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (E.B.); (A.O.)
- Institute of Human Morphology, 117418 Moscow, Russia
| | - Tatyana Popkova
- V. A. Nasonova Institute of Rheumatology, 115522 Moscow, Russia;
| | - Alexander Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (E.B.); (A.O.)
- Institute of Human Morphology, 117418 Moscow, Russia
| |
Collapse
|
18
|
Blocking Aerobic Glycolysis by Targeting Pyruvate Dehydrogenase Kinase in Combination with EGFR TKI and Ionizing Radiation Increases Therapeutic Effect in Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13050941. [PMID: 33668151 PMCID: PMC7956357 DOI: 10.3390/cancers13050941] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Non-small cell lung cancer (NSCLC) patients harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) inevitably develop resistance to targeted EGFR tyrosine kinase inhibitors (TKI) therapy. To support malignant features associated with cancer development and therapy resistance, the cancer cells adapt their metabolic rate and pathways. As an example, aerobic glycolysis, where the cells use glycolysis in the presence of oxygen, is frequently seen. Here we show that targeting aerobic glycolysis represents a promising strategy in cancer therapeutics. Abstract Increased glycolytic activity is a hallmark of cancer initiation and progression and is often observed in non-small cell lung cancer (NSCLC). Pyruvate dehydrogenase (PDH) complex acts as a gatekeeper between glycolysis and oxidative phosphorylation, and activation of PDH is known to inhibit glycolytic activity. As part of a standard therapeutic regimen, patients with NSCLC harboring oncogenic mutations in the epidermal growth factor receptor (EGFR) are treated with EGFR tyrosine kinase inhibitors (EGFR TKIs). Independent of good initial response, development of resistance to this therapy is inevitable. In the presented work, we propose that inhibition of glycolysis will add to the therapeutic effects and possibly prevent development of resistance against both EGFR TKIs and ionizing radiation in NSCLC. Analysis of transcriptome data from two independent NSCLC patient cohorts identified increased expression of pyruvate dehydrogenase kinase 1 (PDHK1) as well as upregulated expression of genes involved in glucose metabolism in tumors compared to normal tissue. We established in vitro models of development of resistance to EGFR TKIs to study metabolism and determine if targeting PDHK would prevent development of resistance to EGFR TKIs in NSCLC cells. The PDHK1 inhibitor dichloroacetate (DCA) in combination with EGFR TKIs and/or ionizing radiation was shown to increase the therapeutic effect in our NSCLC cell models. This mechanism was associated with redirected metabolism towards pyruvate oxidation and reduced lactate production, both in EGFR TKI sensitive and resistant NSCLC cells. Using DCA, the intracellular pool of pyruvate available for lactic fermentation becomes limited. Consequently, pyruvate is redirected to the mitochondria, and reinforces mitochondrial activity. Addition of DCA to cell culture deacidifies the extracellular microenvironment as less lactate is produced and excreted. In our study, we find that this redirection of metabolism adds to the therapeutic effect of EGFR TKI and ionizing radiation in NSCLC.
Collapse
|
19
|
Polat IH, Tarrado-Castellarnau M, Bharat R, Perarnau J, Benito A, Cortés R, Sabatier P, Cascante M. Oxidative Pentose Phosphate Pathway Enzyme 6-Phosphogluconate Dehydrogenase Plays a Key Role in Breast Cancer Metabolism. BIOLOGY 2021; 10:85. [PMID: 33498665 PMCID: PMC7911610 DOI: 10.3390/biology10020085] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
The pentose phosphate pathway (PPP) plays an essential role in the metabolism of breast cancer cells for the management of oxidative stress and the synthesis of nucleotides. 6-phosphogluconate dehydrogenase (6PGD) is one of the key enzymes of the oxidative branch of PPP and is involved in nucleotide biosynthesis and redox maintenance status. Here, we aimed to analyze the functional importance of 6PGD in a breast cancer cell model. Inhibition of 6PGD in MCF7 reduced cell proliferation and showed a significant decrease in glucose consumption and an increase in glutamine consumption, resulting in an important alteration in the metabolism of these cells. No difference in reactive oxygen species (ROS) production levels was observed after 6PGD inhibition, indicating that 6PGD, in contrast to glucose 6-phosphate dehydrogenase, is not involved in redox balance. We found that 6PGD inhibition also altered the stem cell characteristics and mammosphere formation capabilities of MCF7 cells, opening new avenues to prevent cancer recurrance after surgery or chemotherapy. Moreover, inhibition of 6PGD via chemical inhibitor S3 resulted in an induction of senescence, which, together with the cell cycle arrest and apoptosis induction, might be orchestrated by p53 activation. Therefore, we postulate 6PGD as a novel therapeutic target to treat breast cancer.
Collapse
Affiliation(s)
- Ibrahim H. Polat
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Equipe Environnement et Prédiction de la Santé des Populations, Laboratoire TIMC (UMR 5525), CHU de Grenoble, Université Grenoble Alpes, 38700 CEDEX La Tronche, France;
- Department of Medicine, Hematology/Oncology, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Míriam Tarrado-Castellarnau
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28001 Madrid, Spain
| | - Rohit Bharat
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Jordi Perarnau
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Adrian Benito
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Division of Cancer, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Roldán Cortés
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
| | - Philippe Sabatier
- Equipe Environnement et Prédiction de la Santé des Populations, Laboratoire TIMC (UMR 5525), CHU de Grenoble, Université Grenoble Alpes, 38700 CEDEX La Tronche, France;
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain; (I.H.P.); (M.T.-C.); (R.B.); (J.P.); (A.B.); (R.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28001 Madrid, Spain
| |
Collapse
|
20
|
Targeting Endothelial Cell Metabolism by Inhibition of Pyruvate Dehydrogenase Kinase and Glutaminase-1. J Clin Med 2020; 9:jcm9103308. [PMID: 33076309 PMCID: PMC7602423 DOI: 10.3390/jcm9103308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Targeting endothelial cell (EC) metabolism should impair angiogenesis, regardless of how many angiogenic signals are present. The dependency of proliferating ECs on glucose and glutamine for energy and biomass production opens new opportunities for anti-angiogenic therapy in cancer. The aim of the present study was to investigate the role of pyruvate dehydrogenase kinase (PDK) inhibition with dichloroacetate (DCA), alone or in combination with the glutaminase-1 (GLS-1) inhibitor, Bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl) ethyl sulfide (BPTES), on Human umbilical vein endothelial cells (HUVECs) metabolism, proliferation, apoptosis, migration, and vessel formation. We demonstrated that both drugs normalize HUVECs metabolism by decreasing glycolysis for DCA and by reducing glutamate production for BPTES. DCA and BPTES reduced HUVECs proliferation and migration but have no impact on tube formation. While DCA increased HUVECs respiration, BPTES decreased it. Using both drugs in combination further reduced HUVECs proliferation while normalizing respiration and apoptosis induction. Overall, we demonstrated that DCA, a metabolic drug under study to target cancer cells metabolism, also affects tumor angiogenesis. Combining DCA and BPTES may reduce adverse effect of each drug alone and favor tumor angiogenesis normalization.
Collapse
|
21
|
Gan J, Li S, Meng Y, Liao Y, Jiang M, Qi L, Li Y, Bai Y. The influence of photodynamic therapy on the Warburg effect in esophageal cancer cells. Lasers Med Sci 2020; 35:1741-1750. [PMID: 32034563 DOI: 10.1007/s10103-020-02966-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Abstract
To investigate whether the Warburg effect is a key modulator on the resistance mechanism of photodynamic therapy (PDT). Glycolysis was examined by the test of lactate product and glucose uptake at different post-PDT time points. Cell viability was detected by the CCK-8 assay and cell proliferation was detected by colony formation assay. The expression of glycolysis and related proteins were examined by western blotting. Target gene was silenced by RNAi. In the present study, we assessed the effect of PDT on cancer cell glycolysis. Our team has demonstrated that pyruvate kinase M2 (PKM2), a key speed-limiting enzyme of glycolysis, was significantly overexpressed in patients with esophageal cancer. Our results in the present study showed that PKM2 was downregulated, and lactate product and glucose uptake were inhibited in cells exposed to 5-aminolevulinic acid (5-ALA)-mediated PDT at 4 h after treatment. However, at 24 h after PDT, we observed a substantial increase in PKM2 expression, lactate product, and glucose uptake. Moreover, silencing of PKM2 gene abrogated the upregulatory effect of PDT on glycolysis at late post-PDT period. 2-Deoxy-D-glucose (2-DG) is a recognized chemical inhibitor of glycolysis. The combined treatment of 2-DG and PDT significantly inhibited tumor growth in vitro at 24 h. These results demonstrate that PDT drives the Warburg effect in a time-dependent manner, and PKM2 plays an important role in this progress, which indicated that PKM2 may be a potential molecular target to increase the sensitivity of esophageal cancer cells to PDT.
Collapse
Affiliation(s)
- Junqing Gan
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Shumin Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yu Meng
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yuanyu Liao
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Mingxia Jiang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Ling Qi
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yanjing Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
22
|
Sousa B, Pereira J, Marques R, Grilo LF, Pereira SP, Sardão VA, Schmitt F, Oliveira PJ, Paredes J. P-cadherin induces anoikis-resistance of matrix-detached breast cancer cells by promoting pentose phosphate pathway and decreasing oxidative stress. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165964. [PMID: 32920119 DOI: 10.1016/j.bbadis.2020.165964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/27/2022]
Abstract
Successful metastatic spreading relies on cancer cells with stem-like properties, glycolytic metabolism and increased antioxidant protection, allowing them to escape anoikis and to survive in circulation. The expression of P-cadherin, a poor prognostic factor in breast cancer, is associated with hypoxic, glycolytic and acidosis biomarkers. In agreement, P-cadherin-enriched breast cancer cell populations presents a glycolytic and an acid-resistance phenotype. Our aim was to evaluate whether P-cadherin expression controls the glycolytic and oxidative phosphorylation fluxes of matrix-detached breast cancer cells, acting as an antioxidant and enhancing their survival in anchorage-independent conditions. By using matrix-detached breast cancer cells, we concluded that P-cadherin increases glucose-6-phosphate dehydrogenase expression, up-regulating the carbon flux through the pentose phosphate pathway, while inhibiting pyruvate oxidation to acetyl-coA via pyruvate dehydrogenase kinase-4 (PDK-4) activation. Accordingly, P-cadherin expression conferred increased sensitivity to dichloroacetate (DCA), a PDK inhibitor. P-cadherin expression also regulates oxidative stress in matrix-detached breast cancer cells, through the control of antioxidant systems, such as catalase and superoxide dismutases (SOD)1 and 2, providing these cells with an increased resistance to doxorubicin-induced anoikis. Importantly, this association was validated in primary invasive breast carcinomas, where an enrichment of SOD2 was found in P-cadherin-overexpressing breast carcinomas. In conclusion, we propose that P-cadherin up-regulates carbon flux through the pentose phosphate pathway and decreases oxidative stress in matrix-detached breast cancer cells. These metabolic remodeling and antioxidant roles of P-cadherin can promote the survival of breast cancer cells in circulation and in metastatic sites, being a possible player in breast cancer therapeutic resistance to pro-oxidant-based interventions.
Collapse
Affiliation(s)
- Bárbara Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal.
| | - Joana Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal.
| | - Ricardo Marques
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, Portugal
| | - Luís F Grilo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, Portugal
| | - Susana P Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, Portugal.
| | - Fernando Schmitt
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; Medical Faculty of the University of Porto, Porto, Portugal.
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, Portugal.
| | - Joana Paredes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; Medical Faculty of the University of Porto, Porto, Portugal.
| |
Collapse
|
23
|
Reiter RJ, Sharma R, Ma Q, Rorsales-Corral S, de Almeida Chuffa LG. Melatonin inhibits Warburg-dependent cancer by redirecting glucose oxidation to the mitochondria: a mechanistic hypothesis. Cell Mol Life Sci 2020; 77:2527-2542. [PMID: 31970423 PMCID: PMC11104865 DOI: 10.1007/s00018-019-03438-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Melatonin has the ability to intervene in the initiation, progression and metastasis of some experimental cancers. A large variety of potential mechanisms have been advanced to describe the metabolic and molecular events associated with melatonin's interactions with cancer cells. There is one metabolic perturbation that is common to a large number of solid tumors and accounts for the ability of cancer cells to actively proliferate, avoid apoptosis, and readily metastasize, i.e., they use cytosolic aerobic glycolysis (the Warburg effect) to rapidly generate the necessary ATP required for the high metabolic demands of the cancer cells. There are several drugs, referred to as glycolytic agents, that cause cancer cells to abandon aerobic glycolysis and shift to the more conventional mitochondrial oxidative phosphorylation for ATP synthesis as in normal cells. In doing so, glycolytic agents also inhibit cancer growth. Herein, we hypothesize that melatonin also functions as an inhibitor of cytosolic glycolysis in cancer cells using mechanisms, i.e., downregulation of the enzyme (pyruvate dehydrogenase kinase) that interferes with the conversion of pyruvate to acetyl CoA in the mitochondria, as do other glycolytic drugs. In doing so, melatonin halts the proliferative activity of cancer cells, reduces their metastatic potential and causes them to more readily undergo apoptosis. This hypothesis is discussed in relation to the previously published reports. Whereas melatonin is synthesized in the mitochondria of normal cells, we hypothesize that this synthetic capability is not present in cancer cell mitochondria because of the depressed acetyl CoA; acetyl CoA is necessary for the rate limiting enzyme in melatonin synthesis, arylalkylamine-N-acetyltransferase. Finally, the ability of melatonin to switch glucose oxidation from the cytosol to the mitochondria also explains how tumors that become resistant to conventional chemotherapies are re-sensitized to the same treatment when melatonin is applied.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Qiang Ma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Sergio Rorsales-Corral
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | | |
Collapse
|
24
|
Fan Y, Ou L, Fan J, Li L, Wang X, Niu L, Wu X, Luo C. PLCε regulates metabolism and metastasis signaling via HIF-1α/MEK/ERK pathway in prostate cancer. J Cell Physiol 2020; 235:8546-8557. [PMID: 32383180 DOI: 10.1002/jcp.29698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 11/09/2022]
Abstract
Phospholipase C-ε (PLCε) is frequently overexpressed in tumors and plays an important role in the regulation of tumorigenesis. Although great progress has been made in understanding biological roles of PLCε, the relevant molecular mechanisms underlying its pro-tumor activity remain largely unclear. Here, we demonstrated that PLCε knockdown reduced cell metastasis, glucose consumption and lactate production in a manner that depended on hypoxia inducible factor 1α (HIF-1α) expression in prostate cancer cells. Interestingly, our findings showed that the expression levels of PLCε were positively associated with those of HIF-1α in clinical prostate carcinoma samples. Knockdown of PLCε impaired HIF-1α levels and transcriptional activity by regulating the extracellular-signal-regulated kinase pathway, and blocking HIF-1α nuclear translocation. Furthermore, PLCε could interact with the von Hippel-Lindau E3 ligase complex to modulate the stability of HIF-1α. Collectively, our findings demonstrate that PLCε could be a crucial positive regulator of HIF-1α, which would promote PLCε-enhanced tumorigenesis.
Collapse
Affiliation(s)
- Yanru Fan
- Clinical Laboratory, Henan Provincial People's Hospital, Zhengzhou, China
| | - Liping Ou
- Laboratory Medical College, Chongqing Medical University, Chongqing, China
| | - Jiaxin Fan
- Laboratory Medical College, Chongqing Medical University, Chongqing, China
| | - Luo Li
- Laboratory Medical College, Chongqing Medical University, Chongqing, China
| | - Xiao Wang
- First Affliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingfang Niu
- Laboratory Medical College, Chongqing Medical University, Chongqing, China
| | - Xiaohou Wu
- First Affliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunli Luo
- Laboratory Medical College, Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Xiong X, Ke X, Wang L, Yao Z, Guo Y, Zhang X, Chen Y, Pang CP, Schally AV, Zhang H. Splice variant of growth hormone-releasing hormone receptor drives esophageal squamous cell carcinoma conferring a therapeutic target. Proc Natl Acad Sci U S A 2020; 117:6726-6732. [PMID: 32156725 PMCID: PMC7104313 DOI: 10.1073/pnas.1913433117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The extrahypothalamic growth hormone-releasing hormone (GHRH) and its cognate receptors (GHRH-Rs) and splice variants are expressed in a variety of cancers. It has been shown that the pituitary type of GHRH-R (pGHRH-R) mediates the inhibition of tumor growth induced by GHRH-R antagonists. However, GHRH-R antagonists can also suppress some cancers that do not express pGHRH-R, yet the underlying mechanisms have not been determined. Here, using human esophageal squamous cell carcinoma (ESCC) as a model, we were able to reveal that SV1, a known splice variant of GHRH-R, is responsible for the inhibition induced by GHRH-R antagonist MIA-602. We demonstrated that GHRH-R splice variant 1 (SV1) is a hypoxia-driven promoter of tumor progression. Hypoxia-elevated SV1 activates a key glycolytic enzyme, muscle-type phosphofructokinase (PFKM), through the nuclear factor kappa B (NF-κB) pathway, which enhances glycolytic metabolism and promotes progression of ESCC. The malignant actions induced by the SV1-NF-κB-PFKM pathway could be reversed by MIA-602. Altogether, our studies demonstrate a mechanism by which GHRH-R antagonists target SV1. Our findings suggest that SV1 is a hypoxia-induced oncogenic promoter which can be an alternative target of GHRH-R antagonists.
Collapse
Affiliation(s)
- Xiao Xiong
- Department of General Surgery, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, 510632 Guangzhou, Guangdong, China
| | - Xiurong Ke
- Department of Immunotherapy and Gastrointestinal Oncology, Affiliated Cancer Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
- Department of Surgery, Translational Surgical Oncology, University Medical Center Groningen, University of Groningen, GZ 9713 Groningen, The Netherlands
| | - Lu Wang
- Department of General Surgery, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, 510632 Guangzhou, Guangdong, China
| | - Zhimeng Yao
- Department of General Surgery, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, 510632 Guangzhou, Guangdong, China
- Department of Immunotherapy and Gastrointestinal Oncology, Affiliated Cancer Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Yi Guo
- Endoscopy Center, Affiliated Cancer Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Xianyang Zhang
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33136
- South Florida Veterans Affairs Foundation for Research and Education, Veterans Affairs Medical Center, Miami, FL 33136
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Yuping Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, 999077 Hong Kong, China
- Joint Shantou International Eye Center, Shantou University/Chinese University of Hong Kong, 515041 Shantou, China
| | - Andrew V Schally
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33136;
- South Florida Veterans Affairs Foundation for Research and Education, Veterans Affairs Medical Center, Miami, FL 33136
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136
- Division of Medical Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Hao Zhang
- Department of General Surgery, First Affiliated Hospital of Jinan University, 510632 Guangzhou, Guangdong, China;
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, 510632 Guangzhou, Guangdong, China
- Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, China
| |
Collapse
|
26
|
Schoonjans CA, Gallez B. Metabolic Plasticity of Tumor Cells: How They Do Adapt to Food Deprivation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:109-123. [PMID: 32130696 DOI: 10.1007/978-3-030-34025-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulated metabolism is a key hallmark of cancer cells and an enticing target for cancer treatment. Since the last 10 years, research on cancer metabolism has moved from pathway attention to network consideration. This metabolic complexity continuously adapt to new constraints in the tumor microenvironment. In this review, we will highlight striking changes in cancer cell metabolism compared to normal cells. Understanding this tumor metabolic plasticity suggests potential new targets and innovative combinatorial treatments for fighting cancer.
Collapse
Affiliation(s)
- Céline A Schoonjans
- Université catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Brussels, Belgium
| | | |
Collapse
|
27
|
ABA and sucrose co-regulate strawberry fruit ripening and show inhibition of glycolysis. Mol Genet Genomics 2019; 295:421-438. [PMID: 31807909 DOI: 10.1007/s00438-019-01629-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Abscisic acid (ABA) and sucrose play an important role in strawberry fruit ripening, but how ABA and sucrose co-regulate this ripening progress remains unclear. The intention of this study was to examine the effect of ABA and sucrose on strawberry fruit ripening and to evaluate the ABA/sucrose interaction mechanism on the strawberry fruit ripening process. Here, we report that there is an acute synergistic effect between ABA and sucrose in accelerating strawberry fruit ripening. The time frame of fruit development and ripening was shortened after the application of ABA, sucrose, and ABA + sucrose, but most of the major quality parameters in treated-ripe fruit, including fruit weight, total soluble solids, anthocyanin, ascorbic acid, the total phenolic content, lightness (L*), chroma (C*), and hue angle (h°) values were not affected. Meanwhile, the endogenous ABA and sucrose levels, and the expression of ABA and sucrose signaling genes and ripening-related genes, such as NCED1, NCED2, SnRK2.2, SuSy, MYB5, CEL1, and CEL2, was all significantly enhanced by ABA or sucrose treatment alone, but in particular, by the ABA + sucrose treatment. Therefore, improving the ripening regulation efficiency is one synergetic action of ABA/sucrose. Another synergetic action of ABA/sucrose shows that a short inhibition of glycolysis occurs during accelerated strawberry ripening. ABA and sucrose can induce higher accumulation of H2O2, leading to a transient decrease in glycolysis. Conversely, lower endogenous H2O2 levels caused by reduced glutathione (GSH) treatment resulted in a transient increase in glycolysis while delaying strawberry fruit ripening. Collectively, this study demonstrates that the ABA/sucrose interaction affects the ripening regulation efficiency and shows inhibition of glycolysis.
Collapse
|
28
|
Schoonjans CA, Joudiou N, Brusa D, Corbet C, Feron O, Gallez B. Acidosis-induced metabolic reprogramming in tumor cells enhances the anti-proliferative activity of the PDK inhibitor dichloroacetate. Cancer Lett 2019; 470:18-28. [PMID: 31812695 DOI: 10.1016/j.canlet.2019.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/20/2019] [Accepted: 12/01/2019] [Indexed: 01/08/2023]
Abstract
Altered metabolic pathways in cancer such as exacerbated glycolytic flux and increased glutamine metabolism are promising targets for anti-cancer therapies. While commonly observed in glycolytic tumors, extracellular acidosis has never been considered as a potential modulator of anti-metabolic drug activity such as dichloroacetate (DCA). Using cancer cells from various origins selected for their ability to proliferate under acidic conditions, we found that DCA exerts greater inhibitory effects on the growth of these acid-adapted cells than in parental cells. Moreover, daily DCA administration to mice led to a significant decrease in tumor growth from acid-adapted cells but not from parental cells. 13C-tracer studies revealed that DCA induced a double metabolic shift, diminishing glycolysis and increasing intracellular glutamine in acid-adapted cells. As a consequence, DCA reduced the pentose phosphate pathway activity more extensively and increased apoptosis in acid-adapted cells. Finally, the combination of DCA with a glutaminase inhibitor significantly enhanced the cytotoxic effects of DCA. Overall, the interplay between acidosis and DCA exposure leads to metabolic reprogramming that considerably alters cellular fitness.
Collapse
Affiliation(s)
- C A Schoonjans
- Université Catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium; Université Catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology and Therapeutics, Belgium
| | - N Joudiou
- Université Catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Nuclear and Electron Spin Technologies, Brussels, Belgium
| | - D Brusa
- Université Catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique, Flow Cytometry Platform, Belgium
| | - C Corbet
- Université Catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology and Therapeutics, Belgium
| | - O Feron
- Université Catholique de Louvain (UCLouvain), Institut de Recherche Expérimentale et Clinique, Pole of Pharmacology and Therapeutics, Belgium
| | - B Gallez
- Université Catholique de Louvain (UCLouvain), Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium.
| |
Collapse
|
29
|
Duan XL, Ma CC, Hua J, Xiao TW, Luan J. Benzyl butyl phthalate (BBP) triggers the malignancy of acute myeloid leukemia cells via upregulation of PDK4. Toxicol In Vitro 2019; 62:104693. [PMID: 31629899 DOI: 10.1016/j.tiv.2019.104693] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/13/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022]
Abstract
Acute Myeloid Leukemia (AML) is a cancer of hematopoietic stem cells with a rapid progression. Recent studies indicated that endocrine disruptor chemicals (EDCs) are potential risk factors for AML progression. Our present data showed that an industrial endocrine disrupting chemical, Benzyl butyl phthalate (BBP), can promote the proliferation of AML cells and decrease their sensitivity to daunorubicin (DNR) and cytarabine (Ara-C) treatments. Further, BBP can increase the glucose consumption, lactate generation, and ATP levels of AML cells. Among the measured glycolysis-related genes, BBP can increase the expression of pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4), a mitochondrial protein that regulates the tricarboxylic acid cycle (TCA) cycle. The inhibitor of PDK4 or its specific siRNA can attenuate BBP-induced cell proliferation and ATP generation, which suggested the essential roles of PDK4 in BBP-induced glycolysis and proliferation. Further, BBP can increase the mRNA stability of PDK4, while had no effect on its transcription and protein stability. miR-15b-5p can bind with the 3'UTR of PDK4 to decrease its mRNA stability, while BBP can decrease the expression of miR-15b-5p in AML cells. Collectively, our data showed that BBP can trigger the malignancy of AML cells via regulation of miR-15b-5p/PDK4 signals.
Collapse
Affiliation(s)
- Xian-Liang Duan
- Department of Hematology, Liaocheng People's Hospital, Shandong 252000, China
| | - Cong-Cong Ma
- Department of Hematology, Liaocheng People's Hospital, Shandong 252000, China
| | - Jing Hua
- Department of Hematology, Liaocheng People's Hospital, Shandong 252000, China
| | - Tai-Wu Xiao
- Department of Hematology, Liaocheng People's Hospital, Shandong 252000, China
| | - Jing Luan
- Department of Hematology, Liaocheng People's Hospital, Shandong 252000, China.
| |
Collapse
|
30
|
George J, Tuomela T, Kemppainen E, Nurminen A, Braun S, Yalgin C, Jacobs HT. Mitochondrial dysfunction generates a growth-restraining signal linked to pyruvate in Drosophila larvae. Fly (Austin) 2019; 13:12-28. [PMID: 31526131 PMCID: PMC6988875 DOI: 10.1080/19336934.2019.1662266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Drosophila bang-sensitive mutant tko25t, manifesting a global deficiency in oxidative phosphorylation due to a mitochondrial protein synthesis defect, exhibits a pronounced delay in larval development. We previously identified a number of metabolic abnormalities in tko25t larvae, including elevated pyruvate and lactate, and found the larval gut to be a crucial tissue for the regulation of larval growth in the mutant. Here we established that expression of wild-type tko in any of several other tissues of tko25t also partially alleviates developmental delay. The effects appeared to be additive, whilst knockdown of tko in a variety of specific tissues phenocopied tko25t, producing developmental delay and bang-sensitivity. These findings imply the existence of a systemic signal regulating growth in response to mitochondrial dysfunction. Drugs and RNAi-targeted on pyruvate metabolism interacted with tko25t in ways that implicated pyruvate or one of its metabolic derivatives in playing a central role in generating such a signal. RNA-seq revealed that dietary pyruvate-induced changes in transcript representation were mostly non-coherent with those produced by tko25t or high-sugar, consistent with the idea that growth regulation operates primarily at the translational and/or metabolic level.
Collapse
Affiliation(s)
- Jack George
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tea Tuomela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Esko Kemppainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Samuel Braun
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Cagri Yalgin
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Munemoto M, Mukaisho K, Miyashita T, Oyama K, Haba Y, Okamoto K, Kinoshita J, Ninomiya I, Fushida S, Taniura N, Sugihara H, Fujimura T. Roles of the hexosamine biosynthetic pathway and pentose phosphate pathway in bile acid-induced cancer development. Cancer Sci 2019; 110:2408-2420. [PMID: 31215094 PMCID: PMC6676276 DOI: 10.1111/cas.14105] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/17/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022] Open
Abstract
Esophageal squamous cell carcinomas (ESCCs) as well as adenocarcinomas (EACs) were developed in rat duodenal contents reflux models (reflux model). The present study aimed to shed light on the mechanism by which bile acid stimulation causes cancer onset and progression. Metabolomics analyses were performed on samples of neoplastic and nonneoplastic tissues from reflux models, and K14D, cultivated from a nonmetastatic, primary ESCC, and ESCC-DR, established from a metastatic thoracic lesion. ESCC-DRtca2M was prepared by treating ESCC-DR cells with taurocholic acid (TCA) to accelerate cancer progression. The lines were subjected to comprehensive genomic analyses. In addition, protein expression levels of glucose-6-phosphate dehydrogenase (G6PD), nuclear factor kappa B (NF-κB) (p65) and O-linked N-Acetylglucosamine (O-GlcNAc) were compared among lines. Cancers developed in the reflux models exhibited greater hexosamine biosynthesis pathway (HBP) activation compared with the nonneoplastic tissues. Expression of O-GlcNAc transferase (OGT) increased considerably in both ESCC and EAC compared with nonneoplastic squamous epithelium. Conversely, cell line-based experiments revealed the greater activation of the pentose phosphate pathway (PPP) at higher degrees of malignancy. G6PD overexpression in response to TCA exposure was observed. Both NF-κB (p65) and O-GlcNAc were expressed more highly in ESCC-DRtca2M than in the other cell lines. Moreover, ESCC-DRtca2M cells had additional chromosomal abnormalities in excess of ESCC-DR cells. Overall, glucose metabolism was upregulated in both esophageal cancer tissue and cell lines. While bile acids are not mutagenic, chronic exposure seems to trigger NF-κB(p65) activation, potentially inducing genetic mutations as well as facilitating carcinogenesis and cancer progression. Glucose metabolism was upregulated in both esophageal cancer tissue and cell lines, and the HBP was activated in the former. The cell line-based experiments demonstrated upregulation of the pentose phosphate pathway (PPP) at higher degrees of malignancy. While bile acids are not mutagenic, chronic exposure seems to trigger G6PD overexpression and NF-κB (p65) activation, potentially inducing genetic mutations as well as facilitating carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Masayoshi Munemoto
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Ken‐ichi Mukaisho
- Division of Molecular and Diagnostic PathologyDepartment of PathologyShiga University of Medical ScienceOtsuJapan
| | - Tomoharu Miyashita
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Katsunobu Oyama
- Department of SurgeryPublic Central Hospital of Matto IshikawaHakusanJapan
| | - Yusuke Haba
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Koichi Okamoto
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Jun Kinoshita
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Itasu Ninomiya
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Sachio Fushida
- Department of Gastroenterological SurgeryKanazawa University HospitalKanazawaJapan
| | - Naoko Taniura
- Division of Molecular and Diagnostic PathologyDepartment of PathologyShiga University of Medical ScienceOtsuJapan
| | - Hiroyuki Sugihara
- Division of Molecular and Diagnostic PathologyDepartment of PathologyShiga University of Medical ScienceOtsuJapan
| | | |
Collapse
|
32
|
Ralph SJ, Nozuhur S, ALHulais RA, Rodríguez‐Enríquez S, Moreno‐Sánchez R. Repurposing drugs as pro‐oxidant redox modifiers to eliminate cancer stem cells and improve the treatment of advanced stage cancers. Med Res Rev 2019; 39:2397-2426. [DOI: 10.1002/med.21589] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/20/2019] [Accepted: 03/31/2019] [Indexed: 01/10/2023]
Affiliation(s)
- Stephen J. Ralph
- School of Medical ScienceGriffith University Southport Australia
| | - Sam Nozuhur
- School of Medical ScienceGriffith University Southport Australia
| | | | | | | |
Collapse
|
33
|
Spugnini EP, Logozzi M, Di Raimo R, Mizzoni D, Fais S. A Role of Tumor-Released Exosomes in Paracrine Dissemination and Metastasis. Int J Mol Sci 2018; 19:E3968. [PMID: 30544664 PMCID: PMC6321583 DOI: 10.3390/ijms19123968] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
Metastatic diffusion is thought to be a multi-step phenomenon involving the release of cells from the primary tumor and their diffusion through the body. Currently, several hypotheses have been put forward in order to explain the origin of cancer metastasis, including epithelial⁻mesenchymal transition, mutagenesis of stem cells, and a facilitating role of macrophages, involving, for example, transformation or fusion hybridization with neoplastic cells. In this paradigm, tumor-secreted extracellular vesicles (EVs), such as exosomes, play a pivotal role in cell communications, delivering a plethora of biomolecules including proteins, lipids, and nucleic acids. For their natural role in shuttling molecules, EVs have been newly considered a part of the metastatic cascade. They have a prominent role in preparing the so-called "tumor niches" in target organs. However, recent evidence has pointed out an even more interesting role of tumor EVs, consisting in their ability to induce malignant transformation in resident mesenchymal stem cells. All in all, in this review, we discuss the multiple involvements of EVs in the metastatic cascade, and how we can exploit and manipulate EVs in order to reduce the metastatic spread of malignant tumors.
Collapse
Affiliation(s)
| | - Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Rossella Di Raimo
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Davide Mizzoni
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
34
|
Zhou L, Kong Q, Zhang Y, Yang W, Yan S, Li M, Wang Y, Zhou Y, Yu H, Han L. Glucose deprivation promotes apoptotic response to S1 by enhancing autophagy in human cervical cancer cells. Cancer Manag Res 2018; 10:6195-6204. [PMID: 30538566 PMCID: PMC6260140 DOI: 10.2147/cmar.s184180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background S1 is a novel BH3 mimetic that can induce death in some types of cancer cells, such as melanoma B16, ovarian cancer SKOV3, and U251 glioma cells. S1 inhibits Bcl-2 and Mcl-1 expression and induces cancer cell apoptosis. Cancer cell survival is highly dependent on glucose. Here, we observed the effect of glucose deprivation on the apoptotic response to S1 in human cervical cancer (HeLa) cells. Materials and methods Earle’s balanced salt solution (EBSS) was used to simulate glucose deprivation. MTT assay was used to analyze the cell survival rate, and Hoechst 33342 dye was used to detect the apoptosis in HeLa cells. Western blotting was used to detect the expression of ER stress and autophagy relative proteins. In addition, lysosomes were observed with Lyso-Tracker staining by confocal microscopy. Results S1 decreased cell distribution density and survival rate, and MTT assay showed that EBSS enhanced the inhibitory effects of S1 on HeLa cell growth. Hoechst 33342 dye showed that S1 led to pyknosis, fragmentation, and strong staining in HeLa cell nuclei, and EBSS enhanced these effects. Western blotting indicated that EBSS enhanced the expression of apoptosis-related proteins (cytochrome C, caspase-3, and poly[ADP-ribose] polymerase 1) induced by S1 in HeLa cells. S1 decreased p62 expression and increased the autophagosome-associated protein LC3-II expression, which indicated that S1 induced autophagy in HeLa cells. EBSS enhanced S1-induced autophagy in HeLa cells. Furthermore, autophagy inhibitor chloroquine enhanced S1-induced apoptosis in HeLa cells. Conclusion These results indicate that EBSS exacerbates S1-induced autophagy and severe autophagy induced by EBSS and S1 could lead to apoptosis in HeLa cells. The results also suggest that EBSS enhances the sensitivity of HeLa cells to S1-induced apoptosis and that autophagy plays an important role in this process.
Collapse
Affiliation(s)
- Li Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, China, .,Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun 130021, China
| | - Qinghuan Kong
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yunhan Zhang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Wei Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan Yan
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Meihui Li
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yidan Wang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Yanjie Zhou
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Huimei Yu
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun 130021, China,
| | - Liying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130041, China,
| |
Collapse
|
35
|
Eastlack SC, Dong S, Ivan C, Alahari SK. Suppression of PDHX by microRNA-27b deregulates cell metabolism and promotes growth in breast cancer. Mol Cancer 2018; 17:100. [PMID: 30012170 PMCID: PMC6048708 DOI: 10.1186/s12943-018-0851-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/04/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The disruption of normal gene regulation due to microRNA dysfunction is a common event in cancer pathogenesis. MicroRNA-27b is an example of an oncogenic miRNA, and it is frequently upregulated in breast cancer. MicroRNAs have been found to deregulate tumor metabolism, which typically manifests as heightened cellular glucose uptake in consort with increased flux through glycolysis, followed by the preferential conversion of glycolytic pyruvate into lactate (a phenomenon known as the Warburg Effect). Pyruvate Dehydrogenase, an enzyme complex linking glycolysis with downstream oxidative metabolism, represents a key location where regulation of metabolism occurs; PDHX is a key structural component of this complex and is essential for its function. METHODS We sought to characterize the role of miR-27b in breast cancer by identifying novel transcripts under its control. We began by utilizing luciferase, RNA, and protein assays to establish PDHX as a novel target of miR-27b. We then tested whether miR-27b could alter metabolism using several metabolite assay kits and performed a seahorse analysis. We also examined how the altered metabolism might affect cell proliferation. Lastly, we confirmed the relevance of our findings in human breast tumor samples. RESULTS Our data indicate that Pyruvate Dehydrogenase Protein X is a credible target of miR-27b in breast cancer. Mechanistically, by suppressing PDHX, miR-27b altered levels of pyruvate, lactate and citrate, as well as reducing mitochondrial oxidation and promoting extracellular acidification. These changes corresponded with an increased capacity for cell proliferation. In human breast tumor samples, PDHX expression was deficient, and low levels of PDHX were associated with reduced patient survival. CONCLUSIONS MicroRNA-27b targets PDHX, resulting in an altered metabolic configuration that is better suited to fuel biosynthetic processes and cell proliferation, thereby promoting breast cancer progression.
Collapse
Affiliation(s)
- Steven C Eastlack
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, 70112, USA
| | - Shengli Dong
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, 70112, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, LSUHSC School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
36
|
Snyder V, Reed-Newman TC, Arnold L, Thomas SM, Anant S. Cancer Stem Cell Metabolism and Potential Therapeutic Targets. Front Oncol 2018; 8:203. [PMID: 29922594 PMCID: PMC5996058 DOI: 10.3389/fonc.2018.00203] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022] Open
Abstract
Malignant tumors contain heterogeneous populations of cells in various states of proliferation and differentiation. The presence of cancer stem or initiating cells is a well-established concept wherein quiescent and poorly differentiated cells within a tumor mass contribute to drug resistance, and under permissive conditions, are responsible for tumor recurrence and metastasis. A number of studies have identified molecular markers that are characteristic of tissue-specific cancer stem cells (CSCs). Isolation of CSCs has enabled studies on the metabolic status of CSCs. As metabolic plasticity is a hallmark of cancer cell adaptation, the intricacies of CSC metabolism and their phenotypic behavior are critical areas of research. Unlike normal stem cells, which rely heavily on oxidative phosphorylation (OXPHOS) as their primary source of energy, or cancer cells, which are primarily glycolytic, CSCs demonstrate a unique metabolic flexibility. CSCs can switch between OXPHOS and glycolysis in the presence of oxygen to maintain homeostasis and, thereby, promote tumor growth. Here, we review key factors that impact CSC metabolic phenotype including heterogeneity of CSCs across different histologic tumor types, tissue-specific variations, tumor microenvironment, and CSC niche. Furthermore, we discuss how targeting key players of glycolytic and mitochondrial pathways has shown promising results in cancer eradication and attenuation of disease recurrence in preclinical models. In addition, we highlight studies on other potential therapeutic targets including complex interactions within the microenvironment and cellular communications in the CSC niche to interfere with CSC growth, resistance, and metastasis.
Collapse
Affiliation(s)
- Vusala Snyder
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Tamika C Reed-Newman
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Levi Arnold
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States.,Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States.,Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shrikant Anant
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS, United States.,Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
37
|
Icard P, Shulman S, Farhat D, Steyaert JM, Alifano M, Lincet H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist Updat 2018; 38:1-11. [PMID: 29857814 DOI: 10.1016/j.drup.2018.03.001] [Citation(s) in RCA: 389] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/09/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
Cancer cells employ both conventional oxidative metabolism and glycolytic anaerobic metabolism. However, their proliferation is marked by a shift towards increasing glycolytic metabolism even in the presence of O2 (Warburg effect). HIF1, a major hypoxia induced transcription factor, promotes a dissociation between glycolysis and the tricarboxylic acid cycle, a process limiting the efficient production of ATP and citrate which otherwise would arrest glycolysis. The Warburg effect also favors an intracellular alkaline pH which is a driving force in many aspects of cancer cell proliferation (enhancement of glycolysis and cell cycle progression) and of cancer aggressiveness (resistance to various processes including hypoxia, apoptosis, cytotoxic drugs and immune response). This metabolism leads to epigenetic and genetic alterations with the occurrence of multiple new cell phenotypes which enhance cancer cell growth and aggressiveness. In depth understanding of these metabolic changes in cancer cells may lead to the development of novel therapeutic strategies, which when combined with existing cancer treatments, might improve their effectiveness and/or overcome chemoresistance.
Collapse
Affiliation(s)
- Philippe Icard
- Normandie University, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment, BioTICLA axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France; UNICANCER, Comprehensive Cancer Center François Baclesse, BioTICLA lab, Caen, France; Department of Thoracic Surgery, University Hospital of Caen, France
| | | | - Diana Farhat
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), France; Université Lyon Claude Bernard 1, Lyon, France; Department of Chemistry-Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Lebanese University, Faculty of Sciences, Hadath-Beirut, Lebanon
| | - Jean-Marc Steyaert
- Ecole Polytechnique, Laboratoire d'Informatique (LIX), Palaiseau, France
| | - Marco Alifano
- Department of Thoracic Surgery, Paris Center University Hospital, AP-HP, Paris, France; Paris Descartes University, Paris, France
| | - Hubert Lincet
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), France; Université Lyon Claude Bernard 1, Lyon, France; ISPB, Faculté de Pharmacie, Lyon, France.
| |
Collapse
|
38
|
Neveu MA, De Preter G, Joudiou N, Bol A, Brender JR, Saito K, Kishimoto S, Grégoire V, Jordan BF, Krishna MC, Feron O, Gallez B. Multi-modality imaging to assess metabolic response to dichloroacetate treatment in tumor models. Oncotarget 2018; 7:81741-81749. [PMID: 28082726 PMCID: PMC5340254 DOI: 10.18632/oncotarget.13176] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/19/2016] [Indexed: 01/20/2023] Open
Abstract
Reverting glycolytic metabolism is an attractive strategy for cancer therapy as upregulated glycolysis is a hallmark in various cancers. Dichloroacetate (DCA), long used to treat lactic acidosis in various pathologies, has emerged as a promising anti-cancer drug. By inhibiting the pyruvate dehydrogenase kinase, DCA reactivates the mitochondrial function and decreases the glycolytic flux in tumor cells resulting in cell cycle arrest and apoptosis. We recently documented that DCA was able to induce a metabolic switch preferentially in glycolytic cancer cells, leading to a more oxidative phenotype and decreasing proliferation, while oxidative cells remained less sensitive to DCA treatment. To evaluate the relevance of this observation in vivo, the aim of the present study was to characterize the effect of DCA in glycolytic MDA-MB-231 tumors and in oxidative SiHa tumors using advanced pharmacodynamic metabolic biomarkers. Oxygen consumption, studied by 17O magnetic resonance spectroscopy, glucose uptake, evaluated by 18F-FDG PET and pyruvate transformation into lactate, measured using hyperpolarized 13C-magnetic resonance spectroscopy, were monitored before and 24 hours after DCA treatment in tumor bearing mice. In both tumor models, no clear metabolic shift was observed. Surprisingly, all these imaging parameters concur to the conclusion that both glycolytic tumors and oxidative tumors presented a similar response to DCA. These results highlight a major discordance in metabolic cancer cell bioenergetics between in vitro and in vivo setups, indicating critical role of the local microenvironment in tumor metabolic behaviors.
Collapse
Affiliation(s)
- Marie-Aline Neveu
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Géraldine De Preter
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Joudiou
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Anne Bol
- Radiation Oncology Department & Center for Molecular Imaging, Radiotherapy & Oncology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Jeffery R Brender
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Keita Saito
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Shun Kishimoto
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Vincent Grégoire
- Radiation Oncology Department & Center for Molecular Imaging, Radiotherapy & Oncology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Murali C Krishna
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
39
|
Lu W, Wang L, Chen L, Hui S, Rabinowitz JD. Extraction and Quantitation of Nicotinamide Adenine Dinucleotide Redox Cofactors. Antioxid Redox Signal 2018; 28:167-179. [PMID: 28497978 PMCID: PMC5737638 DOI: 10.1089/ars.2017.7014] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIMS Accurate analysis of dinucleotide redox cofactors nicotinamide adenine dinucleotide phosphate reduced (NADPH), nicotinamide adenine dinucleotide phosphate (NADP+), nicotinamide adenine dinucleotide reduced (NADH), and nicotinamide adenine dinucleotide (NAD+) from biological samples is important to understanding cellular redox homeostasis. In this study, we aimed to develop a simple protocol for quenching metabolism and extracting NADPH that avoids interconversion among the reduced forms and the oxidized forms. RESULTS We compared seven different solvents for quenching and extraction of cultured mammalian cells and mouse tissues: a cold aqueous buffer commonly used in enzyme assays with and without detergent, hot aqueous buffer, and cold organic mixtures (80% methanol, buffered 75% acetonitrile, and acidic 40:40:20 acetonitrile:methanol:water with either 0.02 M or 0.1 M formic acid). Extracts were analyzed by liquid chromatography-mass spectrometry (LC-MS). To monitor the metabolite interconversion, cells were grown in 13C6-glucose medium, and unlabeled standards were spiked into the extraction solvents. Interconversion between the oxidized and reduced forms was substantial except for the enzyme assay buffer with detergent, 80% methanol and 40:40:20 acetonitrile:methanol:water, with the 0.1 M formic acid mix giving the least interconversion and best recoveries. Absolute NAD+, NADH, NADP+, and NADPH concentrations in cells and mouse tissues were measured with this approach. INNOVATION We found that the interconversion between the reduced and oxidized forms during extraction is a major barrier to accurately measuring NADPH/NADP+ and NADH/NAD+ ratios. Such interconversion can be monitored by isotope labeling cells and spiking NAD(P)(H) standards. CONCLUSION Extraction with 40:40:20 acetonitrile:methanol:water with 0.1 M formic acid decreases interconversion and, therefore, is suitable for measurement of redox cofactor ratios using LC-MS. This solvent is also useful for general metabolomics. Samples should be neutralized immediately after extraction to avoid acid-catalyzed degradation. When LC-MS is not available and enzyme assays are accordingly used, inclusion of detergent in the aqueous extraction buffer reduces interconversion. Antioxid. Redox Signal. 28, 167-179.
Collapse
Affiliation(s)
- Wenyun Lu
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Lin Wang
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Li Chen
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Sheng Hui
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University , Princeton, New Jersey
| |
Collapse
|
40
|
|
41
|
Wehmas LC, DeAngelo AB, Hester SD, Chorley BN, Carswell G, Olson GR, George MH, Carter JH, Eldridge SR, Fisher A, Vallanat B, Wood CE. Metabolic Disruption Early in Life is Associated With Latent Carcinogenic Activity of Dichloroacetic Acid in Mice. Toxicol Sci 2017; 159:354-365. [PMID: 28962523 PMCID: PMC6223632 DOI: 10.1093/toxsci/kfx146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Early-life environmental factors can influence later-life susceptibility to cancer. Recent evidence suggests that metabolic pathways may mediate this type of latency effect. Previously, we reported that short-term exposure to dichloroacetic acid (DCA) increased liver cancer in mice 84 weeks after exposure was stopped. Here, we evaluated time course dynamics for key events related to this effect. This study followed a stop-exposure design in which 28-day-old male B6C3F1 mice were given the following treatments in drinking water for up to 93 weeks: deionized water (dH2O, control); 3.5 g/l DCA continuously; or 3.5 g/l DCA for 4-52 weeks followed by dH2O. Effects were evaluated at eight interim time points. A short-term biomarker study was used to evaluate DCA effects at 6, 15, and 30 days. Liver tumor incidence was higher in all DCA treatment groups, including carcinomas in 82% of mice previously treated with DCA for only 4 weeks. Direct effects of DCA in the short-term study included decreased liver cell proliferation and marked mRNA changes related to mitochondrial dysfunction and altered cell metabolism. However, all observed short-term effects of DCA were ultimately reversible, and prior DCA treatment did not affect liver cell proliferation, apoptosis, necrosis, or DNA sequence variants with age. Key intermediate events resulting from transient DCA exposure do not fit classical cytotoxic, mitogenic, or genotoxic modes of action for carcinogenesis, suggesting a distinct mechanism associated with early-life metabolic disruption.
Collapse
Affiliation(s)
- Leah C. Wehmas
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Anthony B. DeAngelo
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Susan D. Hester
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Brian N. Chorley
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Gleta Carswell
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Greg R. Olson
- Toxicologic Pathology Associates, Jefferson, AK,
USA
| | - Michael H. George
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | | | | | - Anna Fisher
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Beena Vallanat
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| | - Charles E. Wood
- National Health and Environmental Effects Research
Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC,
USA
| |
Collapse
|
42
|
Pes GM, Bassotti G, Dore MP. Colorectal Cancer Mortality in Relation to Glucose - 6 - Phosphate Dehydrogenase Deficiency and Consanguinity in Sardinia: A Spatial Correlation Analysis. Asian Pac J Cancer Prev 2017; 18:2403-2407. [PMID: 28950694 PMCID: PMC5720643 DOI: 10.22034/apjcp.2017.18.9.2403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most diffuse malignancy in the world. In Southern Europe, the incidence and prevalence are lower than in most Western countries, although some hot spots of increased risk are emerging. In Sardinia, the cancer rate has risen steeply in the last years. Among risk factors for CRC, genomic homozygosity has been postulated. Glucose-6-phosphate dehydrogenase (G6PD) deficiency has been hypothesized to decrease CRC risk. In Sardinians, this disorder has a frequency of 12-24% due to selection by past malaria. In this study the relationship between mortality for CRC, homozygosity and G6PD deficiency was analysed using spatial analysis. Methods: The spatial association between CRC mortality and G6PD deficiency and homozygosity was assessed in the 377 municipalities of the island using ordinary least squares regression and geographically weighted regression. Results: A consanguinity index, available across all municipalities, was used as a proxy for homozygosity. A significant inverse correlation was found between CRC mortality and G6PD deficiency (ρ = ‒0.216; p = 0.002) whereas no association was found for consanguinity (ρ = ‒0.077; p = 0.498). The geographical map of CRC mortality showed a significant clustering in mountain areas compared to the population living in lowlands, whereas hot spot areas of G6PD deficiency were observed on the south-western side of Sardinia. Conclusions: These results indicate that G6PD deficiency might contribute to reduce colon carcinogenesis, and is in line with in vitro and in vivo studies.
Collapse
Affiliation(s)
- Giovanni Mario Pes
- Dipartimento di Medicina Clinica e Sperimentale, University of Sassari, Sassari 07100, Italy.
| | | | | |
Collapse
|
43
|
Inhibition of retinoic acid receptor β signaling confers glycolytic dependence and sensitization to dichloroacetate in melanoma cells. Oncotarget 2017; 8:84210-84223. [PMID: 29137417 PMCID: PMC5663589 DOI: 10.18632/oncotarget.20476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/19/2017] [Indexed: 12/24/2022] Open
Abstract
Dysregulation of metabolism during melanoma progression is tightly associated with the acquisition of genetic and epigenetic alterations in regulators of metabolic pathways. Retinoic acid receptor beta (RARβ) is epigenetically silenced in a large proportion of melanomas, but a link between RARβ and metabolic rewiring of melanoma has not been established. Here, we show that in primary human melanocytes, all-trans retinoic acid (a RARβ agonist) induced growth inhibition accompanied by a decrease in both glycolytic and oxidative metabolism, whereas selective inhibition of RARβ led to an increase in the basal glycolytic rate and increased sensitivity to inhibition of glycolysis. In melanoma cells, inhibition of RARβ promoted lower mitochondrial respiration and higher glycolytic activity, which led to energetic stress and activation of the energy sensor AMP-activated protein kinase. This metabolic shift increased the sensitivity to both glycolytic inhibition and stimulation of mitochondrial metabolism with dichloroacetate, an inhibitor of pyruvate dehydrogenase kinase. In melanoma cells harboring the BRAFV600E mutation, RARβ activation antagonized the effect of the BRAF inhibitor PLX4032 (vemurafenib). Collectively, these data suggest that RARβ signaling is involved in regulating cellular metabolism in melanoma and may provide a potential target in combination treatment strategies.
Collapse
|
44
|
Benfeitas R, Uhlen M, Nielsen J, Mardinoglu A. New Challenges to Study Heterogeneity in Cancer Redox Metabolism. Front Cell Dev Biol 2017; 5:65. [PMID: 28744456 PMCID: PMC5504267 DOI: 10.3389/fcell.2017.00065] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are important pathophysiological molecules involved in vital cellular processes. They are extremely harmful at high concentrations because they promote the generation of radicals and the oxidation of lipids, proteins, and nucleic acids, which can result in apoptosis. An imbalance of ROS and a disturbance of redox homeostasis are now recognized as a hallmark of complex diseases. Considering that ROS levels are significantly increased in cancer cells due to mitochondrial dysfunction, ROS metabolism has been targeted for the development of efficient treatment strategies, and antioxidants are used as potential chemotherapeutic drugs. However, initial ROS-focused clinical trials in which antioxidants were supplemented to patients provided inconsistent results, i.e., improved treatment or increased malignancy. These different outcomes may result from the highly heterogeneous redox responses of tumors in different patients. Hence, population-based treatment strategies are unsuitable and patient-tailored therapeutic approaches are required for the effective treatment of patients. Moreover, due to the crosstalk between ROS, reducing equivalents [e.g., NAD(P)H] and central metabolism, which is heterogeneous in cancer, finding the best therapeutic target requires the consideration of system-wide approaches that are capable of capturing the complex alterations observed in all of the associated pathways. Systems biology and engineering approaches may be employed to overcome these challenges, together with tools developed in personalized medicine. However, ROS- and redox-based therapies have yet to be addressed by these methodologies in the context of disease treatment. Here, we review the role of ROS and their coupled redox partners in tumorigenesis. Specifically, we highlight some of the challenges in understanding the role of hydrogen peroxide (H2O2), one of the most important ROS in pathophysiology in the progression of cancer. We also discuss its interplay with antioxidant defenses, such as the coupled peroxiredoxin/thioredoxin and glutathione/glutathione peroxidase systems, and its reducing equivalent metabolism. Finally, we highlight the need for system-level and patient-tailored approaches to clarify the roles of these systems and identify therapeutic targets through the use of the tools developed in personalized medicine.
Collapse
Affiliation(s)
- Rui Benfeitas
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Jens Nielsen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| |
Collapse
|
45
|
Aucamp J, Bronkhorst AJ, Peters DL, Van Dyk HC, Van der Westhuizen FH, Pretorius PJ. Kinetic analysis, size profiling, and bioenergetic association of DNA released by selected cell lines in vitro. Cell Mol Life Sci 2017; 74:2689-2707. [PMID: 28315952 PMCID: PMC11107759 DOI: 10.1007/s00018-017-2495-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/19/2017] [Accepted: 02/22/2017] [Indexed: 01/09/2023]
Abstract
Although circulating DNA (cirDNA) analysis shows great promise as a screening tool for a wide range of pathologies, numerous stumbling blocks hinder the rapid translation of research to clinical practice. This is related directly to the inherent complexity of the in vivo setting, wherein the influence of complex systems of interconnected cellular responses and putative DNA sources creates a seemingly arbitrary representation of the quantitative and qualitative properties of the cirDNA in the blood of any individual. Therefore, to evaluate the potential of in vitro cell cultures to circumvent the difficulties encountered in in vivo investigations, the purpose of this work was to elucidate the characteristics of the DNA released [cell-free DNA (cfDNA)] by eight different cell lines. This revealed three different forms of cfDNA release patterns and the presence of nucleosomal fragments as well as actively released forms of DNA, which are not only consistently observed in every tested cell line, but also in plasma samples. Correlations between cfDNA release and cellular origin, growth rate, and cancer status were also investigated by screening and comparing bioenergetics flux parameters. These results show statistically significant correlations between cfDNA levels and glycolysis, while no correlations between cfDNA levels and oxidative phosphorylation were observed. Furthermore, several correlations between growth rate, cancer status, and dependency on aerobic glycolysis were observed. Cell cultures can, therefore, successfully serve as closed-circuit models to either replace or be used in conjunction with biofluid samples, which will enable sharper focus on specific cell types or DNA origins.
Collapse
Affiliation(s)
- Janine Aucamp
- Human Metabolomics, North-West University, Hoffman Street, Potchefstroom, 2520, South Africa.
| | - Abel J Bronkhorst
- Human Metabolomics, North-West University, Hoffman Street, Potchefstroom, 2520, South Africa
| | - Dimetrie L Peters
- Human Metabolomics, North-West University, Hoffman Street, Potchefstroom, 2520, South Africa
| | - Hayley C Van Dyk
- Human Metabolomics, North-West University, Hoffman Street, Potchefstroom, 2520, South Africa
| | | | - Piet J Pretorius
- Human Metabolomics, North-West University, Hoffman Street, Potchefstroom, 2520, South Africa
| |
Collapse
|
46
|
Anemone A, Consolino L, Conti L, Reineri F, Cavallo F, Aime S, Longo DL. In vivo evaluation of tumour acidosis for assessing the early metabolic response and onset of resistance to dichloroacetate by using magnetic resonance pH imaging. Int J Oncol 2017; 51:498-506. [DOI: 10.3892/ijo.2017.4029] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/15/2017] [Indexed: 11/05/2022] Open
|
47
|
PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis 2017; 8:e2820. [PMID: 28542136 PMCID: PMC5520749 DOI: 10.1038/cddis.2017.85] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/11/2017] [Accepted: 01/31/2017] [Indexed: 12/21/2022]
Abstract
The p21-activated kinase 4 (PAK4) is overexpressed in different cancers and promotes proliferation of cancer cells. Reprogramming of glucose metabolism is found in most cancer cells which in turn supports rapid proliferation. However, the relationship between PAK4 and glucose metabolism in cancer cells has not been explored. In this study, we reported that PAK4 promoted glucose intake, NADPH production and lipid biosynthesis, leading to an increased proliferation of colon cancer cells. Mechanistically, PAK4 interacted with glucose-6-phosphate dehydrogenase (G6PD), a rate-limiting enzyme of the pentose phosphate pathway and increased G6PD activity via enhancing Mdm2-mediated p53 ubiquitination degradation. In addition, we demonstrated a close positive correlation between PAK4 and G6PD expression in colon cancer specimens. Furthermore, expression of PAK4 or G6PD was positively correlated with an aggressive phenotype of clinical colon cancer. These findings revealed a novel glucose metabolism-related mechanism of PAK4 in promoting colon cancer cell growth, suggesting that PAK4 and/or G6PD blockage might be a potential therapeutic strategy for colon cancer.
Collapse
|
48
|
Lopes-Rodrigues V, Di Luca A, Mleczko J, Meleady P, Henry M, Pesic M, Cabrera D, van Liempd S, Lima RT, O'Connor R, Falcon-Perez JM, Vasconcelos MH. Identification of the metabolic alterations associated with the multidrug resistant phenotype in cancer and their intercellular transfer mediated by extracellular vesicles. Sci Rep 2017; 7:44541. [PMID: 28303926 PMCID: PMC5356019 DOI: 10.1038/srep44541] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/09/2017] [Indexed: 01/02/2023] Open
Abstract
Multidrug resistance (MDR) is a serious obstacle to efficient cancer treatment. Overexpression of P-glycoprotein (P-gp) plays a significant role in MDR. Recent studies proved that targeting cellular metabolism could sensitize MDR cells. In addition, metabolic alterations could affect the extracellular vesicles (EVs) cargo and release. This study aimed to: i) identify metabolic alterations in P-gp overexpressing cells that could be involved in the development of MDR and, ii) identify a potential role for the EVs in the acquisition of the MDR. Two different pairs of MDR and their drug-sensitive counterpart cancer cell lines were used. Our results showed that MDR (P-gp overexpressing) cells have a different metabolic profile from their drug-sensitive counterparts, demonstrating decreases in the pentose phosphate pathway and oxidative phosphorylation rate; increases in glutathione metabolism and glycolysis; and alterations in the methionine/S-adenosylmethionine pathway. Remarkably, EVs from MDR cells were capable of stimulating a metabolic switch in the drug-sensitive cancer cells, towards a MDR phenotype. In conclusion, obtained results contribute to the growing knowledge about metabolic alterations in MDR cells and the role of EVs in the intercellular transfer of MDR. The specific metabolic alterations identified in this study may be further developed as targets for overcoming MDR.
Collapse
Affiliation(s)
- Vanessa Lopes-Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-465 Porto, Portugal.,ICBAS-UP - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4099-003 Porto, Portugal
| | - Alessio Di Luca
- NICB - National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Justyna Mleczko
- Exosomes Laboratory &Metabolomics platform, CIC bioGUNE, CIBERehd, Derio, Spain
| | - Paula Meleady
- NICB - National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Michael Henry
- NICB - National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Milica Pesic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Diana Cabrera
- Exosomes Laboratory &Metabolomics platform, CIC bioGUNE, CIBERehd, Derio, Spain
| | | | - Raquel T Lima
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-465 Porto, Portugal.,Department of Pathology - FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Robert O'Connor
- NICB - National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Juan M Falcon-Perez
- Exosomes Laboratory &Metabolomics platform, CIC bioGUNE, CIBERehd, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-465 Porto, Portugal.,Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
49
|
Danhier P, Bański P, Payen VL, Grasso D, Ippolito L, Sonveaux P, Porporato PE. Cancer metabolism in space and time: Beyond the Warburg effect. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:556-572. [PMID: 28167100 DOI: 10.1016/j.bbabio.2017.02.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/19/2017] [Accepted: 02/02/2017] [Indexed: 02/07/2023]
Abstract
Altered metabolism in cancer cells is pivotal for tumor growth, most notably by providing energy, reducing equivalents and building blocks while several metabolites exert a signaling function promoting tumor growth and progression. A cancer tissue cannot be simply reduced to a bulk of proliferating cells. Tumors are indeed complex and dynamic structures where single cells can heterogeneously perform various biological activities with different metabolic requirements. Because tumors are composed of different types of cells with metabolic activities affected by different spatial and temporal contexts, it is important to address metabolism taking into account cellular and biological heterogeneity. In this review, we describe this heterogeneity also in metabolic fluxes, thus showing the relative contribution of different metabolic activities to tumor progression according to the cellular context. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Pierre Danhier
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium; Louvain Drug Research Institute (LDRI), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 73 box B1.73.08, 1200 Brussels, Belgium
| | - Piotr Bański
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium
| | - Valéry L Payen
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium
| | - Debora Grasso
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, Florence, Italy
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium
| | - Paolo E Porporato
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Avenue Emmanuel Mounier 52 box B1.53.09, 1200 Brussels, Belgium; Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino Italy.
| |
Collapse
|
50
|
Neveu MA, De Preter G, Marchand V, Bol A, Brender JR, Saito K, Kishimoto S, Porporato PE, Sonveaux P, Grégoire V, Feron O, Jordan BF, Krishna MC, Gallez B. Multimodality Imaging Identifies Distinct Metabolic Profiles In Vitro and In Vivo. Neoplasia 2016; 18:742-752. [PMID: 27889643 PMCID: PMC5126136 DOI: 10.1016/j.neo.2016.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 11/24/2022] Open
Abstract
The study of alterations of tumor metabolism should allow the identification of new targets for innovative anticancer strategies. Metabolic alterations are generally established in vitro, and conclusions are often extrapolated to the in vivo situation without further tumor metabolic phenotyping. To highlight the key role of microenvironment on tumor metabolism, we studied the response of glycolytic and oxidative tumor models to metabolic modulations in vitro and in vivo. MDA-MB-231 and SiHa tumor models, characterized in vitro as glycolytic and oxidative, respectively, were studied. Theoretically, when passing from a hypoxic state to an oxygenated state, a Warburg phenotype should conserve a glycolytic metabolism, whereas an oxidative phenotype should switch from glycolytic to oxidative metabolism (Pasteur effect). This challenge was applied in vitro and in vivo to evaluate the impact of different oxic conditions on glucose metabolism. 18F-fluorodeoxyglucose uptake, lactate production, tumor oxygenation, and metabolic fluxes were monitored in vivo using positron emission tomography, microdialysis, electron paramagnetic resonance imaging, and 13C-hyperpolarizated magnetic resonance spectroscopy, respectively. In vitro, MDA-MB-231 cells were glycolytic under both hypoxic and oxygenated conditions, whereas SiHa cells underwent a metabolic shift after reoxygenation. On the contrary, in vivo, the increase in tumor oxygenation (induced by carbogen challenge) led to a similar metabolic shift in glucose metabolism in both tumor models. The major discordance in metabolic patterns observed in vitro and in vivo highlights that any extrapolation of in vitro metabolic profiling to the in vivo situation should be taken cautiously and that metabolic phenotyping using molecular imaging is mandatory in vivo.
Collapse
Affiliation(s)
- Marie-Aline Neveu
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Géraldine De Preter
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Valérie Marchand
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Anne Bol
- Radiation Oncology Department & Center for Molecular Imaging, Radiotherapy & Oncology, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Jeffery R Brender
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Keita Saito
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Shun Kishimoto
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Paolo E Porporato
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Vincent Grégoire
- Radiation Oncology Department & Center for Molecular Imaging, Radiotherapy & Oncology, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium
| | - Murali C Krishna
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|