51
|
Stamatakos S, Beretta GL, Vergani E, Dugo M, Corno C, Corna E, Tinelli S, Frigerio S, Ciusani E, Rodolfo M, Perego P, Gatti L. Deregulated FASN Expression in BRAF Inhibitor-Resistant Melanoma Cells Unveils New Targets for Drug Combinations. Cancers (Basel) 2021; 13:cancers13092284. [PMID: 34068792 PMCID: PMC8126202 DOI: 10.3390/cancers13092284] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolic changes promoting cell survival are involved in metastatic melanoma progression and in the development of drug resistance. In BRAF-inhibitor resistant melanoma cells, we explored the role of FASN, an enzyme involved in lipogenesis overexpressed in metastatic melanoma. Resistant melanoma cells displaying enhanced migratory and pro-invasive abilities increased sensitivity to the BRAF inhibitor PLX4032 upon the molecular targeting of FASN and upon treatment with the FASN inhibitor orlistat. This behavior was associated with a marked apoptosis and caspase 3/7 activation observed for the drug combination. The expression of FASN was found to be inversely associated with drug resistance in BRAF-mutant cell lines, both in a set of six resistant/sensitive matched lines and in the Cancer Cell Line Encyclopedia. A favorable drug interaction in resistant cells was also observed with U18666 A inhibiting DHCR24, which increased upon FASN targeting. The simultaneous combination of the two inhibitors showed a synergistic interaction with PLX4032 in resistant cells. In conclusion, FASN plays a role in BRAF-mutated melanoma progression, thereby creating novel therapeutic opportunities for the treatment of melanoma.
Collapse
Affiliation(s)
- Serena Stamatakos
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Giovanni Luca Beretta
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Elisabetta Vergani
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
| | - Matteo Dugo
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Cristina Corno
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Elisabetta Corna
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Stella Tinelli
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Simona Frigerio
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
| | - Emilio Ciusani
- Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| | - Monica Rodolfo
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (E.V.); (S.F.)
- Correspondence:
| | - Paola Perego
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (S.S.); (G.L.B.); (M.D.); (C.C.); (E.C.); (S.T.); (P.P.)
| | - Laura Gatti
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy;
| |
Collapse
|
52
|
Schiliro C, Firestein BL. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021; 10:cells10051056. [PMID: 33946927 PMCID: PMC8146072 DOI: 10.3390/cells10051056] [Citation(s) in RCA: 291] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells alter metabolic processes to sustain their characteristic uncontrolled growth and proliferation. These metabolic alterations include (1) a shift from oxidative phosphorylation to aerobic glycolysis to support the increased need for ATP, (2) increased glutaminolysis for NADPH regeneration, (3) altered flux through the pentose phosphate pathway and the tricarboxylic acid cycle for macromolecule generation, (4) increased lipid uptake, lipogenesis, and cholesterol synthesis, (5) upregulation of one-carbon metabolism for the production of ATP, NADH/NADPH, nucleotides, and glutathione, (6) altered amino acid metabolism, (7) metabolism-based regulation of apoptosis, and (8) the utilization of alternative substrates, such as lactate and acetate. Altered metabolic flux in cancer is controlled by tumor-host cell interactions, key oncogenes, tumor suppressors, and other regulatory molecules, including non-coding RNAs. Changes to metabolic pathways in cancer are dynamic, exhibit plasticity, and are often dependent on the type of tumor and the tumor microenvironment, leading in a shift of thought from the Warburg Effect and the “reverse Warburg Effect” to metabolic plasticity. Understanding the complex nature of altered flux through these multiple pathways in cancer cells can support the development of new therapies.
Collapse
Affiliation(s)
- Chelsea Schiliro
- Cell and Developmental Biology Graduate Program and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA;
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
- Correspondence: ; Tel.: +1-848-445-8045
| |
Collapse
|
53
|
Metabolic Classification and Intervention Opportunities for Tumor Energy Dysfunction. Metabolites 2021; 11:metabo11050264. [PMID: 33922558 PMCID: PMC8146396 DOI: 10.3390/metabo11050264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
A comprehensive view of cell metabolism provides a new vision of cancer, conceptualized as tissue with cellular-altered metabolism and energetic dysfunction, which can shed light on pathophysiological mechanisms. Cancer is now considered a heterogeneous ecosystem, formed by tumor cells and the microenvironment, which is molecularly, phenotypically, and metabolically reprogrammable. A wealth of evidence confirms metabolic reprogramming activity as the minimum common denominator of cancer, grouping together a wide variety of aberrations that can affect any of the different metabolic pathways involved in cell physiology. This forms the basis for a new proposed classification of cancer according to the altered metabolic pathway(s) and degree of energy dysfunction. Enhanced understanding of the metabolic reprogramming pathways of fatty acids, amino acids, carbohydrates, hypoxia, and acidosis can bring about new therapeutic intervention possibilities from a metabolic perspective of cancer.
Collapse
|
54
|
Tucci J, Chen T, Margulis K, Orgel E, Paszkiewicz RL, Cohen MD, Oberley MJ, Wahhab R, Jones AE, Divakaruni AS, Hsu CC, Noll SE, Sheng X, Zare RN, Mittelman SD. Adipocytes Provide Fatty Acids to Acute Lymphoblastic Leukemia Cells. Front Oncol 2021; 11:665763. [PMID: 33968771 PMCID: PMC8100891 DOI: 10.3389/fonc.2021.665763] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 12/25/2022] Open
Abstract
Background There is increasing evidence that adipocytes play an active role in the cancer microenvironment. We have previously reported that adipocytes interact with acute lymphoblastic leukemia (ALL) cells, contributing to chemotherapy resistance and treatment failure. In the present study, we investigated whether part of this resistance is due to adipocyte provision of lipids to ALL cells. Methods We cultured 3T3-L1 adipocytes, and tested whether ALL cells or ALL-released cytokines induced FFA release. We investigated whether ALL cells took up these FFA, and using fluorescent tagged BODIPY-FFA and lipidomics, evaluated which lipid moieties were being transferred from adipocytes to ALL. We evaluated the effects of adipocyte-derived lipids on ALL cell metabolism using a Seahorse XF analyzer and expression of enzymes important for lipid metabolism, and tested whether these lipids could protect ALL cells from chemotherapy. Finally, we evaluated a panel of lipid synthesis and metabolism inhibitors to determine which were affected by the presence of adipocytes. Results Adipocytes release free fatty acids (FFA) when in the presence of ALL cells. These FFA are taken up by the ALL cells and incorporated into triglycerides and phospholipids. Some of these lipids are stored in lipid droplets, which can be utilized in states of fuel deprivation. Adipocytes preferentially release monounsaturated FFA, and this can be attenuated by inhibiting the desaturating enzyme steroyl-CoA decarboxylase-1 (SCD1). Adipocyte-derived FFA can relieve ALL cell endogenous lipogenesis and reverse the cytotoxicity of pharmacological acetyl-CoA carboxylase (ACC) inhibition. Further, adipocytes alter ALL cell metabolism, shifting them from glucose to FFA oxidation. Interestingly, the unsaturated fatty acid, oleic acid, protects ALL cells from modest concentrations of chemotherapy, such as those that might be present in the ALL microenvironment. In addition, targeting lipid synthesis and metabolism can potentially reverse adipocyte protection of ALL cells. Conclusion These findings uncover a previously unidentified interaction between ALL cells and adipocytes, leading to transfer of FFA for use as a metabolic fuel and macromolecule building block. This interaction may contribute to ALL resistance to chemotherapy, and could potentially be targeted to improve ALL treatment outcome.
Collapse
Affiliation(s)
- Jonathan Tucci
- Diabetes and Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Ting Chen
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA, United States
| | - Katherine Margulis
- Department of Chemistry, Stanford University, Stanford, CA, United States.,The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Etan Orgel
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Rebecca L Paszkiewicz
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA, United States
| | - Michael D Cohen
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA, United States
| | - Matthew J Oberley
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Rachel Wahhab
- Diabetes and Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Anthony E Jones
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA, United States
| | - Ajit S Divakaruni
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA, United States
| | - Cheng-Chih Hsu
- Department of Chemistry, Stanford University, Stanford, CA, United States.,Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Sarah E Noll
- Department of Chemistry, Stanford University, Stanford, CA, United States
| | - Xia Sheng
- Diabetes and Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Richard N Zare
- Department of Chemistry, Stanford University, Stanford, CA, United States
| | - Steven D Mittelman
- Diabetes and Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, United States.,Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, CA, United States
| |
Collapse
|
55
|
Qu H, Shan K, Tang C, Cui G, Fu G, Qi Y, Cui J, Li J, Wang R, Feng N, Chen YQ. A novel small-molecule fatty acid synthase inhibitor with antitumor activity by cell cycle arrest and cell division inhibition. Eur J Med Chem 2021; 219:113407. [PMID: 33901805 DOI: 10.1016/j.ejmech.2021.113407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/13/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
Fatty acid synthase (FASN), the key enzyme in de novo lipogenesis, is an attractive therapeutic target for diseases characterized by excessive lipid accumulation. Many FASN inhibitors have failed in the clinical trial phase, largely because of poor solubility and safety. In this study, we generated a novel small-molecule FASN inhibitor by structure-based virtual screening. PFI09, the lead compound, is easy to synthesize, and inhibits the lipid synthesis in OP9 mammalian cell line and Caenorhabditis elegans as well as the proliferation of several cancer cell lines via the blockade of FASN. Mechanistic investigations show that PFI09 induces S-phase arrest, cell division reduction and apoptosis. We also develop a chemically stable analog of PFI09, MFI03, which reduces the proliferation of PC3 tumor cells both in vitro and in vivo, without toxicity to mice. In summary, our data suggest that MFI03 is an effective FASN inhibitor and a promising antineoplastic drug candidate.
Collapse
Affiliation(s)
- Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Kai Shan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Chunlei Tang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong Province, 519041, China
| | - Guoling Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Yumin Qi
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Jing Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Jiaqi Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Rong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; Department of Urology, Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, 214012, China; Wuxi Clinical College, Nantong University, Wuxi, Jiangsu, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province, 214012, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, 214012, China.
| |
Collapse
|
56
|
Interplay between Metabolism Reprogramming and Epithelial-to-Mesenchymal Transition in Cancer Stem Cells. Cancers (Basel) 2021; 13:cancers13081973. [PMID: 33923958 PMCID: PMC8072988 DOI: 10.3390/cancers13081973] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Tumor cells display important plasticity potential. Notably, tumor cells have the ability to change toward immature cells called cancer stem cells under the influence of the tumor environment. Importantly, cancer stem cells are a small subset of relatively quiescent cells that, unlike rapidly dividing differentiated tumor cells, escape standard chemotherapies, causing relapse or recurrence of cancer. Interestingly, these cells adopt a specific metabolism. Most often, they mainly rely on glucose uptake and metabolism to sustain their energy needs. This metabolic reprogramming is set off by environmental factors such as pro-inflammatory signals or catecholamine hormones (epinephrine, norepinephrine). A better understanding of this process could provide opportunities to kill cancer stem cells. Indeed, it would become possible to develop drugs that act specifically on metabolic pathways used by these cells. These new drugs could be used to strengthen the effects of current chemotherapies and overcome cancers with poor prognoses. Abstract Tumor cells display important plasticity potential, which contributes to intratumoral heterogeneity. Notably, tumor cells have the ability to retrodifferentiate toward immature states under the influence of their microenvironment. Importantly, this phenotypical conversion is paralleled by a metabolic rewiring, and according to the metabostemness theory, metabolic reprogramming represents the first step of epithelial-to-mesenchymal transition (EMT) and acquisition of stemness features. Most cancer stem cells (CSC) adopt a glycolytic phenotype even though cells retain functional mitochondria. Such adaptation is suggested to reduce the production of reactive oxygen species (ROS), protecting CSC from detrimental effects of ROS. CSC may also rely on glutaminolysis or fatty acid metabolism to sustain their energy needs. Besides pro-inflammatory cytokines that are well-known to initiate the retrodifferentiation process, the release of catecholamines in the microenvironment of the tumor can modulate both EMT and metabolic changes in cancer cells through the activation of EMT transcription factors (ZEB1, Snail, or Slug (SNAI2)). Importantly, the acquisition of stem cell properties favors the resistance to standard care chemotherapies. Hence, a better understanding of this process could pave the way for the development of therapies targeting CSC metabolism, providing new strategies to eradicate the whole tumor mass in cancers with unmet needs.
Collapse
|
57
|
Kadri S, El Ayed M, Kadri A, Limam F, Aouani E, Mokni M. Protective effect of grape seed extract and orlistat co-treatment against stroke: Effect on oxidative stress and energy failure. Biomed Pharmacother 2021; 136:111282. [PMID: 33485068 DOI: 10.1016/j.biopha.2021.111282] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Ischemic stroke is a major health concern and a leading cause of mortality worldwide. Oxidative stress is an early event in the course of stroke inducing neuro-inflammation and cell death. Grape seed extract (GSE) is a natural phytochemical mixture exhibiting antioxidant, anti-inflammatory and neuroprotective properties. Orlistat (ORL) is an anti-obesity agent and a gastro-intestinal lipase inhibitor which showed recently beneficial effects on brain lipotoxicity. Recent studies reported the increase of lipase activity upon stroke which led us to investigate the neuroprotective effect of ORL on rat brain I/R injury as well as the putative synergism with GSE. I/R insult infarcted the brain parenchyma as assessed by TTC staining, induced an oxidative stress as revealed by increased lipoperoxidation along with alteration of antioxidant enzymes activities which was corrected using the cotreatment of ORL + GSE. I/R also disturbed the main metabolic pathways involved in brain fueling as glycolysis, neoglucogenesis, glycogenolysis, TCA cycle and electron transfer chain (ETC) complexes. These disturbances were also corrected with the cotreatment ORL + GSE which maintained energetic activities near to the control level. I/R also disrupted transition metals distribution, along with associated enzymes as tyrosinase, LDH or glutamine synthetase activities and induced hippocampal inflammation as revealed by glycogen depletion from dentate gyrus area along with depressed anti-inflammatory IL1β cytokine and increased pro-inflammatory CD68 antigen. Interestingly almost all I/R-induced disturbances were corrected either partially upon ORL and GSE on their own and the best neuroprotection was obtained in the presence of both drugs (ORL + GSE) enabling robust neuroprotection of the sub granular zone within hippocampal dentate gyrus area.
Collapse
Affiliation(s)
- Safwen Kadri
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia.
| | - Mohamed El Ayed
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Amal Kadri
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Ferid Limam
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Ezzedine Aouani
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| | - Meherzia Mokni
- Bioactive Substances Laboratory, Biotechnology Centre, Technopolis Borj-Cedria, BP-901, 2050, Hammam-Lif, Tunisia
| |
Collapse
|
58
|
Shekhawat RS, Mandal CC. Anti-obesity Medications in Cancer Therapy: A Comprehensive Insight. Curr Cancer Drug Targets 2021; 21:476-494. [PMID: 34225630 DOI: 10.2174/1568009621666210322122829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/29/2020] [Accepted: 12/10/2020] [Indexed: 11/22/2022]
Abstract
The interplay between cancer and obesity is multifactorial and complex with the increased risk of cancer development in obese individuals posing a significant threat. Obesity leads to the upregulation or hyperactivation of several oncogenic pathways in cancer cells, which drives them towards a deleterious phenotype. The cross-talk between cancer and obesity is considered a large contributing factor in the development of chemotherapeutic drug resistance and the resistance to radiotherapy. The link between obesity and the development of cancer is so strong that a medication that demonstrates effectiveness against both conditions would serve as an essential step. In this context, anti-obesity medications provide a worthy list of candidates based on their chemo-preventive potential and chemotherapeutic properties. The current study focuses on exploring the potential of anti-obesity medicines as dual anticancer drugs. These medications target several key signaling pathways (e.g., AMPK, PI3K/Akt/mTOR, MAPK, NF-κB, JNK/ERK), which prove to be crucial for both cancer growth and metastases. Some of these drugs also play an important role in attenuating the signaling and cellular events which incite cancer-obesity cross-talk and demonstrate efficient counteraction of neoplastic transformation. Thus, this review highlights a comprehensive view of the potential use of anti-obesity medicines to treat both cancer and obesity for patients exhibiting both comorbities.
Collapse
Affiliation(s)
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, India
| |
Collapse
|
59
|
Zhou J, Zhao J, Su C. Role of Aberrant Lipid Metabolism of Cancer Stem Cells in Cancer Progression. Curr Cancer Drug Targets 2021; 21:631-639. [PMID: 33726650 DOI: 10.2174/1568009619666210316112333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/28/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Cancer stem cells (CSCs) represent a small population of cancer cells that are able to self-renew and initiate tumors, which undergo epigenetic, epithelial-mesenchymal, immunological, and metabolic reprogramming to adapt to the tumor microenvironment as well as survive host defense or therapeutic insults. The metabolic reprogramming that accompanies cancer onset is known to be critical for the disease pathogenesis. A coordinated dysregulation of lipid metabolism is observed in nearly all cancer types. In addition to fulfilling basic requirements of structural lipids for membrane synthesis, lipids function importantly as signaling molecules and contribute to energy homeostasis. In this review, we summarize the current progress in the attractive research field of aberrant lipid metabolism regarding CSCs in cancer progression, which provides insights into therapeutic agents targeting CSCs based upon their modulation of lipid metabolism.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai. China
| | - Jing Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai. China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai. China
| |
Collapse
|
60
|
Schcolnik-Cabrera A, Chavez-Blanco A, Dominguez-Gomez G, Juarez M, Vargas-Castillo A, Ponce-Toledo RI, Lai D, Hua S, Tovar AR, Torres N, Perez-Montiel D, Diaz-Chavez J, Duenas-Gonzalez A. Pharmacological inhibition of tumor anabolism and host catabolism as a cancer therapy. Sci Rep 2021; 11:5222. [PMID: 33664364 PMCID: PMC7933231 DOI: 10.1038/s41598-021-84538-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
The malignant energetic demands are satisfied through glycolysis, glutaminolysis and de novo synthesis of fatty acids, while the host curses with a state of catabolism and systemic inflammation. The concurrent inhibition of both, tumor anabolism and host catabolism, and their effect upon tumor growth and whole animal metabolism, have not been evaluated. We aimed to evaluate in colon cancer cells a combination of six agents directed to block the tumor anabolism (orlistat + lonidamine + DON) and the host catabolism (growth hormone + insulin + indomethacin). Treatment reduced cellular viability, clonogenic capacity and cell cycle progression. These effects were associated with decreased glycolysis and oxidative phosphorylation, leading to a quiescent energetic phenotype, and with an aberrant transcriptomic landscape showing dysregulation in multiple metabolic pathways. The in vivo evaluation revealed a significant tumor volume inhibition, without damage to normal tissues. The six-drug combination preserved lean tissue and decreased fat loss, while the energy expenditure got decreased. Finally, a reduction in gene expression associated with thermogenesis was observed. Our findings demonstrate that the simultaneous use of this six-drug combination has anticancer effects by inducing a quiescent energetic phenotype of cultured cancer cells. Besides, the treatment is well-tolerated in mice and reduces whole animal energetic expenditure and fat loss.
Collapse
Affiliation(s)
- Alejandro Schcolnik-Cabrera
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico
- PECEM, National Autonomous University of Mexico, Mexico City, Mexico
| | - Alma Chavez-Blanco
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico
| | - Guadalupe Dominguez-Gomez
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico
| | - Mandy Juarez
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico
| | - Ariana Vargas-Castillo
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition, Salvador Zubiran, Mexico City, Mexico
| | - Rafael Isaac Ponce-Toledo
- Division of Archaea Biology and Ecogenomics, Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria
| | - Donna Lai
- Molecular Biology Facility, University of Sydney, Sydney, Australia
| | - Sheng Hua
- Molecular Biology Facility, University of Sydney, Sydney, Australia
| | - Armando R Tovar
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition, Salvador Zubiran, Mexico City, Mexico
| | - Nimbe Torres
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition, Salvador Zubiran, Mexico City, Mexico
| | | | - Jose Diaz-Chavez
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico
| | - Alfonso Duenas-Gonzalez
- Division of Basic Research, National Cancer Institute, Ave. San Fernando 22, Tlalpan, 14080, Mexico City, Mexico.
- Unit of Biomedical Research in Cancer, Institute of Biomedical Research, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
61
|
Fu Y, Zou T, Shen X, Nelson PJ, Li J, Wu C, Yang J, Zheng Y, Bruns C, Zhao Y, Qin L, Dong Q. Lipid metabolism in cancer progression and therapeutic strategies. MedComm (Beijing) 2021; 2:27-59. [PMID: 34766135 PMCID: PMC8491217 DOI: 10.1002/mco2.27] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022] Open
Abstract
Dysregulated lipid metabolism represents an important metabolic alteration in cancer. Fatty acids, cholesterol, and phospholipid are the three most prevalent lipids that act as energy producers, signaling molecules, and source material for the biogenesis of cell membranes. The enhanced synthesis, storage, and uptake of lipids contribute to cancer progression. The rewiring of lipid metabolism in cancer has been linked to the activation of oncogenic signaling pathways and cross talk with the tumor microenvironment. The resulting activity favors the survival and proliferation of tumor cells in the harsh conditions within the tumor. Lipid metabolism also plays a vital role in tumor immunogenicity via effects on the function of the noncancer cells within the tumor microenvironment, especially immune-associated cells. Targeting altered lipid metabolism pathways has shown potential as a promising anticancer therapy. Here, we review recent evidence implicating the contribution of lipid metabolic reprogramming in cancer to cancer progression, and discuss the molecular mechanisms underlying lipid metabolism rewiring in cancer, and potential therapeutic strategies directed toward lipid metabolism in cancer. This review sheds new light to fully understanding of the role of lipid metabolic reprogramming in the context of cancer and provides valuable clues on therapeutic strategies targeting lipid metabolism in cancer.
Collapse
Affiliation(s)
- Yan Fu
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Tiantian Zou
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Xiaotian Shen
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Peter J. Nelson
- Medical Clinic and Policlinic IVLudwig‐Maximilian‐University (LMU)MunichGermany
| | - Jiahui Li
- General, Visceral and Cancer SurgeryUniversity Hospital of CologneCologneGermany
| | - Chao Wu
- Department of General Surgery, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jimeng Yang
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Christiane Bruns
- General, Visceral and Cancer SurgeryUniversity Hospital of CologneCologneGermany
| | - Yue Zhao
- General, Visceral and Cancer SurgeryUniversity Hospital of CologneCologneGermany
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| |
Collapse
|
62
|
Abstract
Metabolic reprogramming with heterogeneity is a hallmark of cancer and is at the basis of malignant behaviors. It supports the proliferation and metastasis of tumor cells according to the low nutrition and hypoxic microenvironment. Tumor cells frantically grab energy sources (such as glucose, fatty acids, and glutamine) from different pathways to produce a variety of biomass to meet their material needs via enhanced synthetic pathways, including aerobic glycolysis, glutaminolysis, fatty acid synthesis (FAS), and pentose phosphate pathway (PPP). To survive from stress conditions (e.g., metastasis, irradiation, or chemotherapy), tumor cells have to reprogram their metabolism from biomass production towards the generation of abundant adenosine triphosphate (ATP) and antioxidants. In addition, cancer cells remodel the microenvironment through metabolites, promoting an immunosuppressive microenvironment. Herein, we discuss how the metabolism is reprogrammed in cancer cells and how the tumor microenvironment is educated via the metabolic products. We also highlight potential metabolic targets for cancer therapies.
Collapse
Affiliation(s)
- Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
63
|
Alfhili MA, Hussein HAM, Park Y, Lee MH, Akula SM. Triclosan induces apoptosis in Burkitt lymphoma-derived BJAB cells through caspase and JNK/MAPK pathways. Apoptosis 2021; 26:96-110. [PMID: 33387145 DOI: 10.1007/s10495-020-01650-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2020] [Indexed: 12/23/2022]
Abstract
Burkitt's lymphoma (BL) is the fastest growing human tumor. Current treatment consists of a multiagent regimen of cytotoxic drugs with serious side effjects including tumor lysis, cardiotoxicity, hepatic impairment, neuropathy, myelosuppression, increased susceptibility to malignancy, and death. Furthermore, therapeutic interventions in areas of BL prevalence are not as feasible as in high-income countries. Therefore, there exists an urgent need to identify new therapies with a safer profile and improved accessibility. Triclosan (TCS), an antimicrobial used in personal care products and surgical scrubs, has gained considerable interest as an antitumor agent due to its interference with fatty acid synthesis. Here, we investigate the antitumor properties and associated molecular mechanisms of TCS in BL-derived BJAB cells. Dose-dependent cell death was observed following treatment with 10-100 µM TCS for 24 h, which was associated with membrane phospholipid scrambling, compromised permeability, and cell shrinkage. TCS-induced cell death was accompanied by elevated intracellular calcium, perturbed redox balance, chromatin condensation, and DNA fragmentation. TCS upregulated Bad expression and downregulated that of Bcl2. Moreover, caspase and JNK MAPK signaling were required for the full apoptotic activity of TCS. In conclusion, this report identifies TCS as an antitumor agent and provides new insights into the molecular mechanisms governing TCS-induced apoptosis in BL cells.
Collapse
Affiliation(s)
- Mohammad A Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Hosni A M Hussein
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC. 27834, USA
- Faculty of Science, Assiut Branch, Al Azhar University, Assiut, 71524, Egypt
| | - Youngyong Park
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Myon Hee Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC. 27834, USA.
| |
Collapse
|
64
|
Matsushita Y, Nakagawa H, Koike K. Lipid Metabolism in Oncology: Why It Matters, How to Research, and How to Treat. Cancers (Basel) 2021; 13:474. [PMID: 33530546 PMCID: PMC7865757 DOI: 10.3390/cancers13030474] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Lipids in our body, which are mainly composed of fatty acids, triacylglycerides, sphingolipids, phospholipids, and cholesterol, play important roles at the cellular level. In addition to being energy sources and structural components of biological membranes, several types of lipids serve as signaling molecules or secondary messengers. Metabolic reprogramming has been recognized as a hallmark of cancer, but changes in lipid metabolism in cancer have received less attention compared to glucose or glutamine metabolism. However, recent innovations in mass spectrometry- and chromatography-based lipidomics technologies have increased our understanding of the role of lipids in cancer. Changes in lipid metabolism, so-called "lipid metabolic reprogramming", can affect cellular functions including the cell cycle, proliferation, growth, and differentiation, leading to carcinogenesis. Moreover, interactions between cancer cells and adjacent immune cells through altered lipid metabolism are known to support tumor growth and progression. Characterization of cancer-specific lipid metabolism can be used to identify novel metabolic targets for cancer treatment, and indeed, several clinical trials are currently underway. Thus, we discuss the latest findings on the roles of lipid metabolism in cancer biology and introduce current advances in lipidomics technologies, focusing on their applications in cancer research.
Collapse
Affiliation(s)
| | - Hayato Nakagawa
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; (Y.M.); (K.K.)
| | | |
Collapse
|
65
|
Gan L, Gan Z, Dan Y, Li Y, Zhang P, Chen S, Ye Z, Pan T, Wan C, Hu X, Yu Y. Tetrazanbigen Derivatives as Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) Partial Agonists: Design, Synthesis, Structure-Activity Relationship, and Anticancer Activities. J Med Chem 2021; 64:1018-1036. [PMID: 33423463 DOI: 10.1021/acs.jmedchem.0c01512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tetrazanbigen (TNBG) is a novel sterol isoquinoline derivative with poor water solubility and moderate inhibitory effects on human cancer cell lines via lipoapoptosis induction. Herein, we developed a series of novel TNBG analogues with improved water solubility and antiproliferative activities. The CCK-8 assay enabled us to identify a novel compound, 14g, which strongly inhibited HepG2 and A549 cell growth with IC50 values of 0.54 and 0.47 μM, respectively. The anticancer effects might be explained by the partial activation and upregulation of PPARγ expression, as indicated by the transactivation assay and western blotting evaluation. Furthermore, the in vitro antiproliferative activity was verified in an in vivo xenograft model in which 14g strongly reduced tumor growth at a dose of 10 mg/kg. In line with these positive observations, 14g exhibited an excellent water solubility of 31.4 mg/mL, which was more than 1000-fold higher than that of TNBG (4 μg/mL). Together, these results suggest that 14g is a promising anticancer therapeutic that deserves further investigation.
Collapse
Affiliation(s)
- Linling Gan
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Zongjie Gan
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yanrong Dan
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yaowei Li
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Peiming Zhang
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Shanwen Chen
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Zaijun Ye
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tao Pan
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Chunmei Wan
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xuelian Hu
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Yu Yu
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
66
|
Wang W, Bai L, Li W, Cui J. The Lipid Metabolic Landscape of Cancers and New Therapeutic Perspectives. Front Oncol 2020; 10:605154. [PMID: 33364199 PMCID: PMC7753360 DOI: 10.3389/fonc.2020.605154] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Lipid metabolism reprograming, as a hallmark of malignancy, has received renewed interest in recent years in such areas as energy sources, cell membrane components, and signaling molecules involved in the rapid tumor growth and the adaptation to the tumor microenvironment. Lipid metabolism deregulation in cancer involves multiple aspects, including an increased lipid uptake, endogenous de novo fatty acid synthesis, fatty acid oxidation, and cholesterol accumulation, thereby promoting tumor growth and progression. Recent advances in the understanding of specific metabolic alterations in cancer reveal novel pathogenesis mechanisms and a growing number of drugs targeting lipid metabolism have been applied in anti-tumor therapy. Thus, this review discusses the lipid metabolic landscape of cancers and the interplay with oncogenic signaling, and summarizes potential therapeutic targets to improve the therapeutic efficiency in cancer patients, in order to provide more reference and thinking for the treatment of lipid metabolism of cancer patients.
Collapse
|
67
|
Quantitative proteomics and phosphoproteomic analyses of mouse livers after tick-borne Babesia microti infection. Int J Parasitol 2020; 51:167-182. [PMID: 33242464 DOI: 10.1016/j.ijpara.2020.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022]
Abstract
Babesia microti is a tick-borne protozoan parasite that infects the red blood cells of mice, humans, and other mammals. The liver tissues of BALB/c mice infected with B. microti exhibit severe injury. To further investigate the molecular mechanisms underlying liver injury and liver self-repair after B. microti infection, data-independent acquisition (DIA) quantitative proteomics was used to analyse changes in the expression and phosphorylation of proteins in liver tissues of BALB/c mice during a B. microti infection period and a recovery period. The expression of FABP1 and ACBP, which are related to fatty acid transport in the liver, was downregulated after infection with B. microti, as was the expression of Acox1, Ehhadh and Acaa1a, which are crucial rate-limiting enzymes in the process of fatty acid β oxidation. The phosphorylation levels of AMP-activated protein kinase (AMPK) and Hormone-sensitive lipase (HSL) were also downregulated. In addition, the expression of PSMB9, CTSC, and other immune-related proteins was increased, reflecting an active immune regulation mechanism in the mice. The weights of mice infected with B. microti were significantly reduced, and the phosphorylation levels of IRS-1, c-Raf, mTOR, and other proteins related to growth and development were downregulated.
Collapse
|
68
|
Schcolnik-Cabrera A, Chavez-Blanco A, Dominguez-Gomez G, Juarez M, Lai D, Hua S, Tovar AR, Diaz-Chavez J, Duenas-Gonzalez A. The combination of orlistat, lonidamine and 6-diazo-5-oxo-L-norleucine induces a quiescent energetic phenotype and limits substrate flexibility in colon cancer cells. Oncol Lett 2020; 20:3053-3060. [PMID: 32782623 PMCID: PMC7400019 DOI: 10.3892/ol.2020.11838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer upregulates glycolysis, glutaminolysis and lipogenesis, and induces a catabolic state in patients. The concurrent inhibition of both tumor anabolism and host catabolism, and the energetic consequences of such an approach, have not previously been fully investigated. In the present study, CT26.WT murine colon cancer cells were treated with the combination of anti-anabolic drugs orlistat, lonidamine and 6-diazo-5-oxo-L-norleucine (DON; OLD scheme), which are inhibitors of the de novo synthesis of fatty acids, glycolysis and glutaminolysis, respectively. In addition, the effects of OLD scheme sumplemented with the combination of anti-catabolic compounds, namely growth hormone, insulin and indomethacin (GII scheme), were also evaluated. The effects of the compounds used in combination on CT26.WT cell viability, clonogenicity and energetic metabolism were assessed in vitro. The results demonstrated that the anti-anabolic approach reduced cell viability, clonogenicity and cell cycle progression, and increased apoptosis. These effects were associated with decreased oxidative phosphorylation, glycolysis and fuel flexibility. Furthermore, the anti-catabolic scheme, alone or supplemented with anti-anabolic compounds, did not favor tumor growth. These findings indicated that the simultaneous pharmacological inhibition of tumor anabolism and host catabolism exhibits antitumor effects that should be further evaluated.
Collapse
Affiliation(s)
| | - Alma Chavez-Blanco
- Division of Basic Research, National Cancer Institute, Mexico City 14080, Mexico
| | | | - Mandy Juarez
- Division of Basic Research, National Cancer Institute, Mexico City 14080, Mexico
| | - Donna Lai
- Molecular Biology Facility, University of Sydney, Sydney, NSW 2006, Australia
| | - Sheng Hua
- Molecular Biology Facility, University of Sydney, Sydney, NSW 2006, Australia
| | - Armando R. Tovar
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico City 14080, Mexico
| | - Jose Diaz-Chavez
- Division of Basic Research, National Cancer Institute, Mexico City 14080, Mexico
| | - Alfonso Duenas-Gonzalez
- Division of Basic Research, National Cancer Institute, Mexico City 14080, Mexico
- Unit of Biomedical Research in Cancer, Institute of Biomedical Research, National Autonomous University of Mexico, Mexico City 14080, Mexico
| |
Collapse
|
69
|
Dilly AK, Honick BD, Lee YJ, Bartlett DL, Choudry HA. Synergistic apoptosis following endoplasmic reticulum stress aggravation in mucinous colon cancer. Orphanet J Rare Dis 2020; 15:211. [PMID: 32811515 PMCID: PMC7437176 DOI: 10.1186/s13023-020-01499-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mucinous colon cancers (MCC) are characterized by abundant production of mucin 2 (MUC2) protein and are less sensitive to standard systemic chemotherapy. We postulated that severe/persistent endoplasmic reticulum stress (ERS) aggravation in MCC would overwhelm compensatory cytoprotective pathways and induce apoptosis. RESULTS Basal levels of ERS markers were higher in MCC and dnTCF-LS174T cells than non-mucinous tumors and these levels were significantly increased by combinatorial treatment with ERS aggravators celecoxib + orlistat. Combination treatment inhibited cell viability and synergistically induced apoptosis. Treatment-induced cell death was ERS-dependent, apoptotic pathways were not activated following knockdown of ERS protein CHOP. Dual drug treatment significantly reduced mucinous tumor growth in vivo and induced ERS and apoptosis, consistent with in vitro experiments. CONCLUSIONS Novel therapies are needed since MCC are more resistant to standard systemic chemotherapy. This study suggests ERS aggravation is a viable therapeutic strategy to reduce tumor growth in MCC.
Collapse
Affiliation(s)
- Ashok K Dilly
- Department of Surgery, University of Pittsburgh Medical Center, Hillman Cancer Center, 5150 Centre Avenue, Suite 414, Pittsburgh, PA, 15232, USA
| | - Brendon D Honick
- Department of Surgery, University of Pittsburgh Medical Center, Hillman Cancer Center, 5150 Centre Avenue, Suite 414, Pittsburgh, PA, 15232, USA
| | - Yong J Lee
- Department of Surgery, University of Pittsburgh Medical Center, Hillman Cancer Center, 5150 Centre Avenue, Suite 414, Pittsburgh, PA, 15232, USA
| | - David L Bartlett
- Department of Surgery, University of Pittsburgh Medical Center, Hillman Cancer Center, 5150 Centre Avenue, Suite 414, Pittsburgh, PA, 15232, USA
| | - Haroon A Choudry
- Department of Surgery, University of Pittsburgh Medical Center, Hillman Cancer Center, 5150 Centre Avenue, Suite 414, Pittsburgh, PA, 15232, USA.
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
70
|
Montesdeoca N, López M, Ariza X, Herrero L, Makowski K. Inhibitors of lipogenic enzymes as a potential therapy against cancer. FASEB J 2020; 34:11355-11381. [PMID: 32761847 DOI: 10.1096/fj.202000705r] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 01/05/2023]
Abstract
Cancer cells rely on several metabolic pathways such as lipid metabolism to meet the increase in energy demand, cell division, and growth and successfully adapt to challenging environments. Fatty acid synthesis is therefore commonly enhanced in many cancer cell lines. Thus, relevant efforts are being made by the scientific community to inhibit the enzymes involved in lipid metabolism to disrupt cancer cell proliferation. We review the rapidly expanding body of inhibitors that target lipid metabolism, their side effects, and current status in clinical trials as potential therapeutic approaches against cancer. We focus on their molecular, biochemical and structural properties, selectivity and effectiveness and discuss their potential role as antitumor drugs.
Collapse
Affiliation(s)
- Nicolás Montesdeoca
- School of Chemical Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
| | - Marta López
- School of Chemical Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
| | - Xavier Ariza
- Department of Inorganic and Organic Chemistry, School of Chemistry, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Kamil Makowski
- School of Chemical Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
| |
Collapse
|
71
|
Weighed Gene Coexpression Network Analysis Screens the Potential Long Noncoding RNAs and Genes Associated with Progression of Coronary Artery Disease. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:8183420. [PMID: 32695216 PMCID: PMC7361886 DOI: 10.1155/2020/8183420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Background Coronary artery disease (CAD) is a type of heart disease with a high morbidity rate. This study is aimed at identifying potential biomarkers closely related to the progression of CAD. Materials and Methods A microarray dataset of GSE59867 was downloaded from a public database, Gene Expression Omnibus, which included 46 cases of stable CAD without a history of myocardial infarction (MI), 30 cases of MI without heart failure (HF), and 34 cases of MI with HF. Differentially expressed long noncoding RNAs (DElncRNAs) and mRNAs (DEmRNAs) were identified by the limma package, and functions of DEmRNAs were annotated by Gene Ontology and KEGG pathways. In addition, weighed gene coexpression network analysis (WGCNA) was used to construct a coexpression network of DEmRNAs, and a disease-related lncRNAs-mRNAs-pathway network was constructed. Finally, the datasets of GSE61145 and GSE57338 were used to verify the expression levels of the above highly correlated candidates. Results A total of 2362 upregulated mRNAs and 2816 downregulated mRNAs, as well as 235 upregulated lncRNAs and 113 downregulated lncRNAs were screened. These genes were significantly enriched in “cytokine-cytokine receptor interaction,” “RIG-I-like receptor signaling pathway,” and “natural killer cell-mediated cytotoxicity.” Five modules including 1201 DEmRNAs were enriched in WGCNA. A coexpression network including 19 DElncRNAs and 413 DEmRNAs was constructed. These genes were significantly enriched in “phosphatidylinositol signaling system,” “insulin signaling pathway,” and “MAPK signaling pathway”. Disease-related gene-pathway network suggested FASN in “insulin signaling pathway,” DGKZ in “phosphatidylinositol signaling system,” and TNFRSF1A in “MAPK signaling pathway” were involved in MI. Conclusion FASN, DGKZ, and TNFRSF1A were revealed to be CAD progression-associated genes by WGCNA coexpression network analysis.
Collapse
|
72
|
Butler LM, Perone Y, Dehairs J, Lupien LE, de Laat V, Talebi A, Loda M, Kinlaw WB, Swinnen JV. Lipids and cancer: Emerging roles in pathogenesis, diagnosis and therapeutic intervention. Adv Drug Deliv Rev 2020; 159:245-293. [PMID: 32711004 PMCID: PMC7736102 DOI: 10.1016/j.addr.2020.07.013] [Citation(s) in RCA: 365] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
With the advent of effective tools to study lipids, including mass spectrometry-based lipidomics, lipids are emerging as central players in cancer biology. Lipids function as essential building blocks for membranes, serve as fuel to drive energy-demanding processes and play a key role as signaling molecules and as regulators of numerous cellular functions. Not unexpectedly, cancer cells, as well as other cell types in the tumor microenvironment, exploit various ways to acquire lipids and extensively rewire their metabolism as part of a plastic and context-dependent metabolic reprogramming that is driven by both oncogenic and environmental cues. The resulting changes in the fate and composition of lipids help cancer cells to thrive in a changing microenvironment by supporting key oncogenic functions and cancer hallmarks, including cellular energetics, promoting feedforward oncogenic signaling, resisting oxidative and other stresses, regulating intercellular communication and immune responses. Supported by the close connection between altered lipid metabolism and the pathogenic process, specific lipid profiles are emerging as unique disease biomarkers, with diagnostic, prognostic and predictive potential. Multiple preclinical studies illustrate the translational promise of exploiting lipid metabolism in cancer, and critically, have shown context dependent actionable vulnerabilities that can be rationally targeted, particularly in combinatorial approaches. Moreover, lipids themselves can be used as membrane disrupting agents or as key components of nanocarriers of various therapeutics. With a number of preclinical compounds and strategies that are approaching clinical trials, we are at the doorstep of exploiting a hitherto underappreciated hallmark of cancer and promising target in the oncologist's strategy to combat cancer.
Collapse
Affiliation(s)
- Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, SA 5005, Australia; South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Ylenia Perone
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London, UK
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Leslie E Lupien
- Program in Experimental and Molecular Medicine, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 037560, USA
| | - Vincent de Laat
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Massimo Loda
- Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - William B Kinlaw
- The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium.
| |
Collapse
|
73
|
Silvas JA, Jureka AS, Nicolini AM, Chvatal SA, Basler CF. Inhibitors of VPS34 and lipid metabolism suppress SARS-CoV-2 replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.18.210211. [PMID: 32743584 PMCID: PMC7386504 DOI: 10.1101/2020.07.18.210211] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutics targeting replication of SARS coronavirus 2 (SARS-CoV-2) are urgently needed. Coronaviruses rely on host membranes for entry, establishment of replication centers and egress. Compounds targeting cellular membrane biology and lipid biosynthetic pathways have previously shown promise as antivirals and are actively being pursued as treatments for other conditions. Here, we tested small molecule inhibitors that target membrane dynamics or lipid metabolism. Included were inhibitors of the PI3 kinase VPS34, which functions in autophagy, endocytosis and other processes; Orlistat, an inhibitor of lipases and fatty acid synthetase, is approved by the FDA as a treatment for obesity; and Triacsin C which inhibits long chain fatty acyl-CoA synthetases. VPS34 inhibitors, Orlistat and Triacsin C inhibited virus growth in Vero E6 cells and in the human airway epithelial cell line Calu-3, acting at a post-entry step in the virus replication cycle. Of these the VPS34 inhibitors exhibit the most potent activity.
Collapse
Affiliation(s)
- Jesus A. Silvas
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303
- Equal contribution
| | - Alexander S. Jureka
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303
- Equal contribution
| | | | | | - Christopher F. Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303
| |
Collapse
|
74
|
Hao P, Xiong Y, Wu H, Yang Y. Network Pharmacology Research and Preliminary Verification of Gegen Qinlian Decoction for the Treatment of Non-Alcoholic Fatty Liver Disease. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20920023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Gegen Qinlian decoction (GQD) is a traditional Chinese medicine that is used to treat non-alcoholic fatty liver disease (NAFLD) in the clinic. The pharmacodynamics and cellular pathways governing the effects of GQD on NAFLD remain undefined. In this study, we investigated GQD pharmacology through assessment of its chemical constituents and evaluated and screened its components using drug likeness, pharmacokinetic characteristics (absorption, distribution, metabolism, excretion, and toxicity), and appropriate compensation mechanisms. We performed predictions of the active GQD ingredients based on reverse pharmacophore matching and compared multiple NAFLD-related genes to determine potential GQD targets. Molecular docking experiments of the active components were performed to reveal cellular targets. Annotation analysis of both target genes and related pathways were assessed through the DAVID database. Cytoscape software was used to construct a “component-target-path” network for the treatment of NAFLD by GQD. Through data analysis, 9 active GQD substances and 10 targets related to NAFLD encompassing 4 cellular pathways were identified. Data were verified through enzyme-linked immunosorbent assay and Western blot analysis. These findings provide new references for the network pharmacology of Chinese medicinal compounds and NAFLD treatment.
Collapse
Affiliation(s)
- Pengfei Hao
- Hubei University of Chinese Medicine, Wuhan, China
- Nanyang Institute of Technology, Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang, China
| | - Yiyi Xiong
- Hubei University of Chinese Medicine, Wuhan, China
| | - Hezhen Wu
- Hubei University of Chinese Medicine, Wuhan, China
| | - Yanfang Yang
- Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
75
|
Orlistat delays hepatocarcinogenesis in mice with hepatic co-activation of AKT and c-Met. Toxicol Appl Pharmacol 2020; 392:114918. [DOI: 10.1016/j.taap.2020.114918] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022]
|
76
|
Evaluation of the antiviral activity of orlistat (tetrahydrolipstatin) against dengue virus, Japanese encephalitis virus, Zika virus and chikungunya virus. Sci Rep 2020; 10:1499. [PMID: 32001767 PMCID: PMC6992670 DOI: 10.1038/s41598-020-58468-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/15/2020] [Indexed: 11/20/2022] Open
Abstract
Many mosquito transmitted viruses of the genera Alphavirus and Flavivirus are human pathogens of significant concern, and there is currently no specific antiviral for any member of these two genera. This study sought to investigate the broad utility of orlistat (tetrahydrolipstatin) in reducing virus infection for several mosquito borne viruses including flaviviruses (dengue virus (DENV; nine isolates analyzed), Japanese encephalitis virus (JEV; one isolate analyzed) and Zika virus (ZIKV; 2 isolates analyzed)) as well as an alphavirus (chikungunya virus; CHIKV; 2 isolates analyzed). Three different treatment regimens were evaluated, namely pre-treatment (only), post-treatment (only) and pre- and post-treatment, and three factors were evaluated, namely level of infection, virus titer and genome copy number. Results showed that all three treatment modalities were able to significantly reduce virus titer for all viruses investigated, with the exception of three isolates of DENV in the pre-treatment only regimen. Pre- and post-treatment was more effective in reducing the level of infection and genome copy number of all viruses investigated than either pre-treatment or post-treatment alone. Collectively, these results suggest orlistat has potential as a broad-spectrum agent against multiple mosquito transmitted viruses.
Collapse
|
77
|
Bueno MJ, Quintela-Fandino M. Emerging role of Fatty acid synthase in tumor initiation: implications for cancer prevention. Mol Cell Oncol 2020; 7:1709389. [PMID: 32158923 PMCID: PMC7051128 DOI: 10.1080/23723556.2019.1709389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 11/24/2022]
Abstract
Targeting metabolic reprogramming has emerged as a promising strategy for therapeutic intervention in cancer. We identify that fatty acid synthase (FASN) is essential for cancer initiation playing a critical role in acquiring three-dimensional (3D) growth properties during transformation. In vivo inhibition of FASN before oncogenic activation prevents tumor development and invasive growth suggesting that FASN could be a potential target for cancer prevention.
Collapse
Affiliation(s)
- Maria J Bueno
- Breast Cancer Clinical Research Unit, CNIO-Spanish National Cancer Research Center, Madrid, Spain
| | - Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit, CNIO-Spanish National Cancer Research Center, Madrid, Spain.,Medical Oncology, Hospital Universitario Quiron, Pozuelo de Alarcon, Madrid, Spain.,Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Madrid, Spain
| |
Collapse
|
78
|
Crocetin Extracted from Saffron Shows Antitumor Effects in Models of Human Glioblastoma. Int J Mol Sci 2020; 21:ijms21020423. [PMID: 31936544 PMCID: PMC7013996 DOI: 10.3390/ijms21020423] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022] Open
Abstract
Over recent years, many authors discussed the effects of different natural compounds on glioblastoma (GBM). Due to its capacity to impair survival and progression of different cancer types, saffron extract (SE), named crocetin (CCT), is particularly noteworthy. In this work, we elucidated the antitumor properties of crocetin in glioma in vivo and in vitro models for the first time. The in vitro results showed that the four tumor cell lines observed in this study (U251, U87, U138, and U373), which were treated with increasing doses of crocetin, showed antiproliferative and pro-differentiative effects as demonstrated by a significant reduction in the number of viable cells, deep changes in cell morphology, and the modulation of mesenchymal and neuronal markers. Indeed, crocetin decreased the expression of Cluster of Differentiation CD44, CD90, CXCR4, and OCT3/4 mesenchymal markers, but increased the expression of βIII-Tubulin and neurofilaments (NFH) neuronal linage-related markers. Epigenetic mechanisms may modulate these changes, since Histone Deacetylase, HDAC1 and HDAC3 were downmodulated in U251 and U87 cells, whereas HDAC1 expression was downmodulated in U138 and U373 cells. Western blotting analyses of Fatty Acid Synthase, FASN, and CD44 resulted in effective inhibition of these markers after CCT treatment, which was associated with important activation of the apoptosis program and reduced glioma cell movement and wound repair. The in vivo studies aligned with the results obtained in vitro. Indeed, crocetin was demonstrated to inhibit the growth of U251 and U87 cells that were subcutaneously injected into animal models. In particular, the Tumor To Progression or TTP values and Kaplan-Meier curves indicated that crocetin had more major effects than radiotherapy alone, but similar effects to temozolomide (TMZ). An intra-brain cell inoculation of a small number of luciferase-transfected U251 cells provided a model that was able to recapitulate recurrence after surgical tumor removal. The results obtained from the orthotopic intra-brain model indicated that CCT treatment increased the disease-free survival (DFS) and overall survival (OS) rates, inducing a delay in appearance of a detectable bioluminescent lesion. CCT showed greater efficacy than Radio Therapy (RT) but comparable efficacy to temozolomide in xenograft models. Therefore, we aimed to continue the study of crocetin's effects in glioma disease, focusing our attention on the radiosensitizing properties of the natural compound and highlighting the ways in which this was realized.
Collapse
|
79
|
Gonzalez-Fierro A, Dueñas-González A. Drug repurposing for cancer therapy, easier said than done. Semin Cancer Biol 2019; 68:123-131. [PMID: 31877340 DOI: 10.1016/j.semcancer.2019.12.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/26/2019] [Accepted: 12/15/2019] [Indexed: 12/24/2022]
Abstract
Drug repurposing for cancer therapy is currently a hot topic of research. Theoretically, in contrast to the known hurdles of developing new molecular entities, the approach of repurposing has several advantages. Mostly, it is said that it is faster, safer, easier, and cheaper. In the real world, however, there are only three repurposed drugs so far, that are listed in widely recognized cancer guidelines, but a large number of them are being studied. Among the many barriers to repurposing cancer drugs, economical-driven are the most important that difficult the clinical development of them. In this review, we provide an overview of the current status of drug repurposing for cancer therapy and the barriers that need to be overcome to realize the benefit of this approach. It means to have repositioned drugs for cancer therapy accepted as standard therapy for cancer indications at low cost.
Collapse
Affiliation(s)
| | - Alfonso Dueñas-González
- Division of Basic Researach, Instituto Nacional de Cancerología, Mexico City, Mexico; Unit of Biomedical Research in Cancer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autonoma de Mexico NAM/ Instituto Nacional de Cancerología, Mexico City, Mexico.
| |
Collapse
|
80
|
Cioccoloni G, Aquino A, Notarnicola M, Caruso MG, Bonmassar E, Zonfrillo M, Caporali S, Faraoni I, Villivà C, Fuggetta MP, Franzese O. Fatty acid synthase inhibitor orlistat impairs cell growth and down-regulates PD-L1 expression of a human T-cell leukemia line. J Chemother 2019; 32:30-40. [PMID: 31775585 DOI: 10.1080/1120009x.2019.1694761] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fatty Acid Synthase (FASN) is responsible for the de novo synthesis of fatty acids, which are involved in the preservation of biological membrane structure, energy storage and assembly of factors involved in signal transduction. FASN plays a critical role in supporting tumor cell growth, thus representing a potential target for anti-cancer therapies. Moreover, this enzyme has been recently associated with increased PD-L1 expression, suggesting a role for fatty acids in the impairment of the immune response in the tumor microenvironment. Orlistat, a tetrahydrolipstatin used for the treatment of obesity, has been reported to reduce FASN activity, while inducing a sensible reduction of the growth potential in different cancer models. We have analyzed the effect of orlistat on different features involved in the tumor cell biology of the T-ALL Jurkat cell line. In particular, we have observed that orlistat inhibits Jurkat cell growth and induces a perturbation of cell cycle along with a decline of FASN activity and protein levels. Moreover, the drug produces a remarkable impairment of PD-L1 expression. These findings suggest that orlistat interferes with different mechanisms involved in the control of tumor cell growth and can potentially contribute to decrease the tumor-associated immune-pathogenesis.
Collapse
Affiliation(s)
- Giorgia Cioccoloni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angelo Aquino
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases S. de Bellis, Bari, Italy
| | - Maria Gabriella Caruso
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases S. de Bellis, Bari, Italy
| | - Enzo Bonmassar
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Manuela Zonfrillo
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Simona Caporali
- Laboratory of Molecular Oncology, IDI-IRCCS Rome, Rome, Italy
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Cristina Villivà
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Maria Pia Fuggetta
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
81
|
Fatty acid synthase and stearoyl-CoA desaturase-1 are conserved druggable cofactors of Old World Alphavirus genome replication. Antiviral Res 2019; 172:104642. [PMID: 31678479 DOI: 10.1016/j.antiviral.2019.104642] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 01/12/2023]
Abstract
Chikungunya virus (CHIKV) is a rapidly emerging mosquito-borne RNA virus that causes epidemics of debilitating disease in tropical and sub-tropical regions with autochtonous transmission in regions with temperate climate. Currently, there is no licensed vaccine or specific antiviral drug available against CHIKV infection. In this study, we examine the role, in the CHIKV viral cycle, of fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD1), two key lipogenic enzymes required for fatty acid production and early desaturation. We show that both enzymes and their upstream regulator PI3K are required for optimal CHIKV infection. We demonstrate that pharmacologic manipulation of FASN or SCD1 enzymatic activity by non-toxic concentrations of cerulenin or CAY10566 decreases CHIKV genome replication. Interestingly, a similar inhibitory effect was also obtained with Orlistat, an FDA-approved anti-obesity drug that targets FASN activity. These drugs were also effective against Mayaro virus (MAYV), an under-studied arthritogenic Old world Alphavirus endemic in South American countries with potential risk of emergence, urbanization and dispersion to other regions. Altogether, our results identify FASN and SCD1 as conserved druggable cofactors of Alphavirus genome replication and support the broad-spectrum activity of drugs targeting the host fatty acids metabolism.
Collapse
|
82
|
Wishart DS. Metabolomics for Investigating Physiological and Pathophysiological Processes. Physiol Rev 2019; 99:1819-1875. [PMID: 31434538 DOI: 10.1152/physrev.00035.2018] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
83
|
Bokil A, Sancho P. Mitochondrial determinants of chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:634-646. [PMID: 35582564 PMCID: PMC8992520 DOI: 10.20517/cdr.2019.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
Chemoresistance constitute nowadays the major contributor to therapy failure in most cancers. There are main factors that mitigate cell response to therapy, such as target organ, inherent sensitivity to the administered compound, its metabolism, drug efflux and influx or alterations on specific cellular targets, among others. We now know that intrinsic properties of cancer cells, including metabolic features, substantially contribute to chemoresistance. In fact, during the last years, numerous reports indicate that cancer cells resistant to chemotherapy demonstrate significant alterations in mitochondrial metabolism, membrane polarization and mass. Metabolic activity and expression of several mitochondrial proteins are modulated under treatment to cope with stress, making these organelles central players in the development of resistance to therapies. Here, we review the role of mitochondria in chemoresistant cells in terms of metabolic rewiring and function of key mitochondria-related proteins.
Collapse
Affiliation(s)
- Ansooya Bokil
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| | - Patricia Sancho
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| |
Collapse
|
84
|
Fatty Acid Inhibition Sensitizes Androgen-Dependent and -Independent Prostate Cancer to Radiotherapy via FASN/NF-κB Pathway. Sci Rep 2019; 9:13284. [PMID: 31527721 PMCID: PMC6746859 DOI: 10.1038/s41598-019-49486-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022] Open
Abstract
Elevated fatty acid synthase (FASN) has been reported in both androgen-dependent and -independent prostate cancers. Conventional treatment for prostate cancer is radiotherapy (RT); however, the following radiation-induced radioresistance often causes treatment failure. Upstream proteins of FASN such as Akt and NF-κB are found increased in the radioresistant prostate cancer cells. Nevertheless, whether inhibition of FASN could improve RT outcomes and reverse radiosensitivity of prostate cancer cells is still unknown. Here, we hypothesised that orlistat, a FASN inhibitor, could improve RT outcomes in prostate cancer. Orlistat treatment significantly reduced the S phase population in both androgen-dependent and -independent prostate cancer cells. Combination of orlistat and RT significantly decreased NF-κB activity and related downstream proteins in both prostate cancer cells. Combination effect of orlistat and RT was further investigated in both LNCaP and PC3 tumour-bearing mice. Combination treatment showed the best tumour inhibition compared to that of orlistat alone or RT alone. These results suggest that prostate cancer treated by conventional RT could be improved by orlistat via inhibition of FASN.
Collapse
|
85
|
Montrose DC, Galluzzi L. Drugging cancer metabolism: Expectations vs. reality. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 347:1-26. [PMID: 31451211 DOI: 10.1016/bs.ircmb.2019.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As compared to their normal counterparts, neoplastic cells exhibit a variety of metabolic changes that reflect not only genetic and epigenetic defects underlying malignant transformation, but also the nutritional and immunobiological conditions of the tumor microenvironment. Such alterations, including the so-called Warburg effect (an increase in glucose uptake largely feeding anabolic and antioxidant metabolism), have attracted considerable attention as potential targets for the development of novel anticancer therapeutics. However, very few drugs specifically conceived to target bioenergetic cancer metabolism are currently approved by regulatory agencies for use in humans. This reflects the elevated degree of heterogeneity and redundancy in the metabolic circuitries exploited by neoplastic cells from different tumors (even of the same type), as well as the resemblance of such metabolic pathways to those employed by highly proliferating normal cells. Here, we summarize the major metabolic alterations that accompany oncogenesis, the potential of targeting bioenergetic metabolism for cancer therapy, and the obstacles that still prevent the clinical translation of such a promising therapeutic paradigm.
Collapse
Affiliation(s)
- David C Montrose
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
86
|
Yadav S, Pandey SK, Goel Y, Temre MK, Singh SM. Diverse Stakeholders of Tumor Metabolism: An Appraisal of the Emerging Approach of Multifaceted Metabolic Targeting by 3-Bromopyruvate. Front Pharmacol 2019; 10:728. [PMID: 31333455 PMCID: PMC6620530 DOI: 10.3389/fphar.2019.00728] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Malignant cells possess a unique metabolic machinery to endure unobstructed cell survival. It comprises several levels of metabolic networking consisting of 1) upregulated expression of membrane-associated transporter proteins, facilitating unhindered uptake of substrates; 2) upregulated metabolic pathways for efficient substrate utilization; 3) pH and redox homeostasis, conducive for driving metabolism; 4) tumor metabolism-dependent reconstitution of tumor growth promoting the external environment; 5) upregulated expression of receptors and signaling mediators; and 6) distinctive genetic and regulatory makeup to generate and sustain rearranged metabolism. This feat is achieved by a "battery of molecular patrons," which acts in a highly cohesive and mutually coordinated manner to bestow immortality to neoplastic cells. Consequently, it is necessary to develop a multitargeted therapeutic approach to achieve a formidable inhibition of the diverse arrays of tumor metabolism. Among the emerging agents capable of such multifaceted targeting of tumor metabolism, an alkylating agent designated as 3-bromopyruvate (3-BP) has gained immense research focus because of its broad spectrum and specific antineoplastic action. Inhibitory effects of 3-BP are imparted on a variety of metabolic target molecules, including transporters, metabolic enzymes, and several other crucial stakeholders of tumor metabolism. Moreover, 3-BP ushers a reconstitution of the tumor microenvironment, a reversal of tumor acidosis, and recuperative action on vital organs and systems of the tumor-bearing host. Studies have been conducted to identify targets of 3-BP and its derivatives and characterization of target binding for further optimization. This review presents a brief and comprehensive discussion about the current state of knowledge concerning various aspects of tumor metabolism and explores the prospects of 3-BP as a safe and effective antineoplastic agent.
Collapse
Affiliation(s)
| | | | | | | | - Sukh Mahendra Singh
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
87
|
Zhang ZG, Zhang HS, Sun HL, Liu HY, Liu MY, Zhou Z. KDM5B promotes breast cancer cell proliferation and migration via AMPK-mediated lipid metabolism reprogramming. Exp Cell Res 2019; 379:182-190. [DOI: 10.1016/j.yexcr.2019.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/24/2019] [Accepted: 04/05/2019] [Indexed: 12/24/2022]
|
88
|
Chang L, Fang S, Chen Y, Yang Z, Yuan Y, Zhang J, Ye L, Gu W. Inhibition of FASN suppresses the malignant biological behavior of non-small cell lung cancer cells via deregulating glucose metabolism and AKT/ERK pathway. Lipids Health Dis 2019; 18:118. [PMID: 31122252 PMCID: PMC6533754 DOI: 10.1186/s12944-019-1058-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022] Open
Abstract
Background Fatty acid synthase (FASN) is overexpressed in most human carcinomas, including non-small cell lung cancer (NSCLC), and contributes to poor prognosis. An increasing number of studies have highlighted the potential function of FASN as both a biomarker and therapeutic target for cancers. However, the underlying molecular mechanisms of FASN in glucose metabolism and the malignant biological behavior of NSCLC remain the subjects of intensive investigation. Methods FASN expression was depleted by FASN-siRNA in A549 and NCI-H1299 cell lines to detect the function of glucose metabolism and the malignant biological behavior of NSCLC cells. Western-blot and qPCR were applied to determine the expressions of FASN, t-AKT, p-AKT, t-ERK, p-ERK, PKM2, HK2 and AZGP1. ATP and lactate were detected to determine the activation of glucose metabolism. CCK8 and transwell assays were used to detect the proliferation, invasion, and migration capacity of the two types of NSCLC cells. The xenograft mouse model was used to evaluate tumor weights after suppression of FASN. Results LV-FASN-siRNA and its control lentiviral vector were successfully transfected into the two types of NSCLC cells (A549 and NCI-H1299). LV-FASN siRNA significantly suppressed FASN expression in both NSCLC cell types, and expressions of p-AKT, p-ERK, PKM2, and AZGP1 were also significantly decreased. Notably, the levels of ATP and lactate were significantly decreased after transfection with LV-FASN siRNA. The proliferation of both NSCLC cell types was decreased after suppression of FASN. The invasion and migration capacity of A549, but not NCI-H1299, were inhibited following down-regulation of FASN. In vivo, inhibition of FASN caused a marked animal tumor weight loss. Conclusions FASN was involved in glucose metabolism via down-regulation of the AKT/ERK pathway and eventually altered the malignant phenotype in lung cancer cells.
Collapse
Affiliation(s)
- Ligong Chang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Surong Fang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Yubao Chen
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Zhenhua Yang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Yuan Yuan
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Jing Zhang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China
| | - Liang Ye
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China.
| | - Wei Gu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210001, People's Republic of China.
| |
Collapse
|