1
|
Akasaka Y, Hasei S, Ohata Y, Kanna M, Nakatsu Y, Sakoda H, Fujishiro M, Kushiyama A, Ono H, Matsubara A, Hinata N, Asano T, Yamamotoya T. Auraptene Enhances AMP-Activated Protein Kinase Phosphorylation and Thereby Inhibits the Proliferation, Migration and Expression of Androgen Receptors and Prostate-Specific Antigens in Prostate Cancer Cells. Int J Mol Sci 2023; 24:16011. [PMID: 37958994 PMCID: PMC10650886 DOI: 10.3390/ijms242116011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Citrus hassaku extract reportedly activates AMPK. Because this extract contains an abundance of auraptene, we investigated whether pure auraptene activates AMPK and inhibits proliferation using prostate cancer cell lines. Indeed, auraptene inhibited the proliferation and migration of LNCaP cells and induced phosphorylation of AMPK or its downstream ACC in LNCaP, PC3, and HEK-293 cells, but not in DU145 cells not expressing LKB1. In addition, the mTOR-S6K pathway, located downstream from activated AMPK, was also markedly suppressed by auraptene treatment. Importantly, it was shown that auraptene reduced androgen receptor (AR) and prostate-specific antigen (PSA) expressions at both the protein and the mRNA level. This auraptene-induced downregulation of PSA was partially but significantly reversed by treatment with AMPK siRNA or the AMPK inhibitor compound C, suggesting AMPK activation to, at least partially, be causative. Finally, in DU145 cells lacking the LKB1 gene, exogenously induced LKB1 expression restored AMPK phosphorylation by auraptene, indicating the essential role of LKB1. In summary, auraptene is a potent AMPK activator that acts by elevating the AMP/ATP ratio, thereby potentially suppressing prostate cancer progression, via at least three molecular mechanisms, including suppression of the mTOR-S6K pathway, reduced lipid synthesis, and AR downregulation caused by AMPK activation.
Collapse
Affiliation(s)
- Yasuyuki Akasaka
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Shun Hasei
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yukino Ohata
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Machi Kanna
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yusuke Nakatsu
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Hideyuki Sakoda
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Midori Fujishiro
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Akifumi Kushiyama
- Department of Pharmacotherapy, Meiji Pharmaceutical University, Kiyose 204-8588, Japan
| | - Hiraku Ono
- Department of Clinical Cell Biology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Akio Matsubara
- Department of Urology, JA Hiroshima General Hospital, Hatsukaichi 738-8503, Japan
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Tomoichiro Asano
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takeshi Yamamotoya
- Department of Biomedical Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
2
|
Mallick R, Bhowmik P, Duttaroy AK. Targeting fatty acid uptake and metabolism in cancer cells: A promising strategy for cancer treatment. Biomed Pharmacother 2023; 167:115591. [PMID: 37774669 DOI: 10.1016/j.biopha.2023.115591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
Despite scientific development, cancer is still a fatal disease. The development of cancer is thought to be significantly influenced by fatty acids. Several mechanisms that control fatty acid absorption and metabolism are reported to be altered in cancer cells to support their survival. Cancer cells can use de novo synthesis or uptake of extracellular fatty acid if one method is restricted. This factor makes it more difficult to target one pathway while failing to treat the disease properly. Side effects may also arise if several inhibitors simultaneously target many targets. If a viable inhibitor could work on several routes, the number of negative effects might be reduced. Comparative investigations against cell viability have found several potent natural and manmade substances. In this review, we discuss the complex roles that fatty acids play in the development of tumors and the progression of cancer, newly discovered and potentially effective natural and synthetic compounds that block the uptake and metabolism of fatty acids, the adverse side effects that can occur when multiple inhibitors are used to treat cancer, and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Prasenjit Bhowmik
- Department of Chemistry, Uppsala Biomedical Centre, Uppsala University, Sweden
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| |
Collapse
|
3
|
Lin E, Garmo H, Hagström E, Van Hemelrijck M, Adolfsson J, Stattin P, Zethelius B, Crawley D. Association between atherogenic lipids and GnRH agonists for prostate cancer in men with T2DM: a nationwide, population-based cohort study in Sweden. Br J Cancer 2023; 128:814-824. [PMID: 36522475 PMCID: PMC9977763 DOI: 10.1038/s41416-022-02091-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Gonadotropin-releasing hormone agonists (GnRH) used in prostate cancer (PCa) are associated with atherogenic dyslipidaemia. It can be assumed that GnRH need to be used with greater caution in men with type 2 diabetes mellitus (T2DM). This study investigated association of GnRH with atherogenic lipids (AL) in PCa men with T2DM. METHODS Two cohorts including 38,311 men with 11 years follow-up based on Swedish national registers were defined (PCa-Exposure cohort and GnRH-Exposure cohort). Based on European guidelines on cardiovascular diseases (CVD), primary outcomes were defined as: 1.0 mmol/L increase in AL and lipid-lowering therapy (LLT) intensification. We used Cox proportional-hazards models and Kaplan-Meier curves to assess the association. RESULTS There was an association between GnRH and increased AL (i.e., triglyceride, PCa-Exposure cohort: HR 1.77, 95% CI 1.48-2.10; GnRH-Exposure cohort: HR 1.88, 95% CI 1.38-2.57). There was also an association between PCa diagnosis and increased AL. In contrast, no association between LLT intensification and GnRH was found. CONCLUSION In this large population-based study, men with T2DM on GnRH for PCa had an increased risk of increased atherogenic lipids. These results highlight the need to closely monitor lipids and to be ready to intensify lipid-lowering therapy in men with T2DM on GnRH for PCa.
Collapse
Affiliation(s)
- E Lin
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King's College London, London, UK.
| | - Hans Garmo
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King's College London, London, UK.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden.,Uppsala Clinical Research Centre, Uppsala, Sweden
| | - Mieke Van Hemelrijck
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King's College London, London, UK
| | - Jan Adolfsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Björn Zethelius
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Danielle Crawley
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King's College London, London, UK
| |
Collapse
|
4
|
Tan SK, Hougen HY, Merchan JR, Gonzalgo ML, Welford SM. Fatty acid metabolism reprogramming in ccRCC: mechanisms and potential targets. Nat Rev Urol 2023; 20:48-60. [PMID: 36192502 PMCID: PMC10826284 DOI: 10.1038/s41585-022-00654-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
Lipid droplet formation is a defining histological feature in clear-cell renal cell carcinoma (ccRCC) but the underlying mechanisms and importance of this biological behaviour have remained enigmatic. De novo fatty acid (FA) synthesis, uptake and suppression of FA oxidation have all been shown to contribute to lipid storage, which is a necessary tumour adaptation rather than a bystander effect. Clinical studies and mechanistic investigations into the roles of different enzymes in FA metabolism pathways have revealed new metabolic vulnerabilities that hold promise for clinical effect. Several metabolic alterations are associated with worse clinical outcomes in patients with ccRCC, as lipogenic genes drive tumorigenesis. Enzymes involved in the intrinsic FA metabolism pathway include FA synthase, acetyl-CoA carboxylase, ATP citrate lyase, stearoyl-CoA desaturase 1, cluster of differentiation 36, carnitine palmitoyltransferase 1A and the perilipin family, and each might be potential therapeutic targets in ccRCC owing to the link between lipid deposition and ccRCC risk. Adipokines and lipid species are potential biomarkers for diagnosis and treatment monitoring in patients with ccRCC. FA metabolism could potentially be targeted for therapeutic intervention in ccRCC as small-molecule inhibitors targeting the pathway have shown promising results in preclinical models.
Collapse
Affiliation(s)
- Sze Kiat Tan
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Helen Y Hougen
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jaime R Merchan
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Mark L Gonzalgo
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Scott M Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
5
|
Fan Y, Lin F, Zhang R, Wang M, Gu R, Long C. Acer truncatum Bunge: A comprehensive review on ethnobotany, phytochemistry and pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114572. [PMID: 34487848 DOI: 10.1016/j.jep.2021.114572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/22/2021] [Accepted: 08/26/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acer truncatum Bunge is a multifunctional plant in northern China. It has traditionally been used to prevent cardiovascular and cerebrovascular diseases and treat skin trauma by different linguistic groups including Mongolian, Tibetan, and Korean. Although research has verified that A. truncatum contains a variety of active ingredients, especially nervonic acid, an important component in delaying brain aging, to date no review has been made to compile its traditional use, phytochemistry, and pharmacology. AIMS OF THE REVIEW This review aimed to update the traditional uses, phytochemistry, and pharmacology of A. truncatum, which expect to provide theoretical support for the future utilization as well as highlight the further investigation of this important plant. MATERIALS AND METHODS The ethnobotanical, phytochemical, and pharmacological information related to A. truncatum from 1949 to March 2021 were collated by surveying the traditional medicinal books and ethnomedicinal publications and searching the online databases including Google Scholar, Sci Finder, Web of Science, Springer Link, PubMed, Wiley, China National Knowledge Infrastructure (CNKI), Baidu Scholar, and Wan Fang Database. RESULTS A. truncatum has traditionally been used for medicinal, edible and ornamental purposes in northern China for many centuries. Different parts of the plant including leaves, fruits and bark, are mainly used as herbal medicine to treat hyperpiesia, hyperlipidemia, bruises, back pain, etc. A total of 288 compounds in A. truncatum, including polyphenols, organic acids or lipids, and biological volatile organic compounds were isolated or identified by phytochemical studies. Pharmacological research showed that A. truncatum has various bioactivities such as acetylcholinesterase inhibition, antibacterial, antioxidant, antitumor, and fatty acid synthase inhibition effects. CONCLUSION A. truncatum has been used as a traditional herbal medicine for centuries in northern China. Polyphenols, organic acids, lipids and other compounds were isolated or identified from different parts of the plant. Most of the pharmacological activities of A. truncatum have been reported, which showed its potential in the development of new drugs or nutraceuticals. However, detailed information on the molecular mechanisms, metabolic activity, and toxicology of active components is limited. Further comprehensive research to evaluate the medicinal properties of A. truncatum will be necessary.
Collapse
Affiliation(s)
- Yanxiao Fan
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| | - Fengke Lin
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| | - Ruifei Zhang
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| | - Miaomiao Wang
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| | - Ronghui Gu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Guizhou University), Ministry of Education, Guiyang, 550025, China; School of Liquor and Food Engineering, Guizhou University, Guiyang, 550025, China.
| | - Chunlin Long
- Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China; Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, 100081, China.
| |
Collapse
|
6
|
Munir R, Lisec J, Swinnen JV, Zaidi N. Too complex to fail? Targeting fatty acid metabolism for cancer therapy. Prog Lipid Res 2021; 85:101143. [PMID: 34856213 DOI: 10.1016/j.plipres.2021.101143] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/19/2022]
Abstract
Given the central role of fatty acids in cancer pathophysiology, the exploitation of fatty acid metabolism as a potential antineoplastic therapy has gained much attention. Several natural and synthetic compounds targeting fatty acid metabolism were hitherto identified, and their effectiveness against cancer cell proliferation and survival was determined. This review will discuss the most clinically viable inhibitors or drugs targeting various proteins or enzymes mapped on nine interconnected fatty acid metabolism-related processes. We will discuss the general significance of each of these processes and the effects of their inhibition on cancer cell progression. Moreover, their mechanisms of action, limitations, and future perspectives will be assessed.
Collapse
Affiliation(s)
- Rimsha Munir
- Cancer Biology Lab, Institute of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan; Hormone Lab Lahore, Pakistan
| | - Jan Lisec
- Bundesanstalt für Materialforschung und -prüfung (BAM), Department of Analytical Chemistry, Richard-Willstätter-Straße 11, 12489 Berlin, Germany
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Nousheen Zaidi
- Cancer Biology Lab, Institute of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan; Cancer Research Center (CRC), University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
7
|
Sena LA, Denmeade SR. Fatty Acid Synthesis in Prostate Cancer: Vulnerability or Epiphenomenon? Cancer Res 2021; 81:4385-4393. [PMID: 34145040 PMCID: PMC8416800 DOI: 10.1158/0008-5472.can-21-1392] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 01/07/2023]
Abstract
Tumor metabolism supports the energetic and biosynthetic needs of rapidly proliferating cancer cells and modifies intra- and intercellular signaling to enhance cancer cell invasion, metastasis, and immune evasion. Prostate cancer exhibits unique metabolism with high rates of de novo fatty acid synthesis driven by activation of the androgen receptor (AR). Increasing evidence suggests that activation of this pathway is functionally important to promote prostate cancer aggressiveness. However, the mechanisms by which fatty acid synthesis are beneficial to prostate cancer have not been well defined. In this review, we summarize evidence indicating that fatty acid synthesis drives progression of prostate cancer. We also explore explanations for this phenomenon and discuss future directions for targeting this pathway for patient benefit.
Collapse
Affiliation(s)
- Laura A Sena
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Ruiz-Pérez MV, Sainero-Alcolado L, Oliynyk G, Matuschek I, Balboni N, Ubhayasekera SKA, Snaebjornsson MT, Makowski K, Aaltonen K, Bexell D, Serra D, Nilsson R, Bergquist J, Schulze A, Arsenian-Henriksson M. Inhibition of fatty acid synthesis induces differentiation and reduces tumor burden in childhood neuroblastoma. iScience 2021; 24:102128. [PMID: 33659885 PMCID: PMC7895756 DOI: 10.1016/j.isci.2021.102128] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/28/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
Many metabolic pathways, including lipid metabolism, are rewired in tumors to support energy and biomass production and to allow adaptation to stressful environments. Neuroblastoma is the second deadliest solid tumor in children. Genetic aberrations, as the amplification of the MYCN-oncogene, correlate strongly with disease progression. Yet, there are only a few molecular targets successfully exploited in the clinic. Here we show that inhibition of fatty acid synthesis led to increased neural differentiation and reduced tumor burden in neuroblastoma xenograft experiments independently of MYCN-status. This was accompanied by reduced levels of the MYCN or c-MYC oncoproteins and activation of ERK signaling. Importantly, the expression levels of genes involved in de novo fatty acid synthesis showed prognostic value for neuroblastoma patients. Our findings demonstrate that inhibition of de novo fatty acid synthesis is a promising pharmacological intervention strategy for the treatment of neuroblastoma independently of MYCN-status.
Collapse
Affiliation(s)
- María Victoria Ruiz-Pérez
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Lourdes Sainero-Alcolado
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Ganna Oliynyk
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Isabell Matuschek
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Nicola Balboni
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - S.J. Kumari A. Ubhayasekera
- Analytical Chemistry, Department of Chemistry and Science for Life Laboratory, Uppsala University, 751 24 Uppsala, Sweden
| | | | - Kamil Makowski
- Department of Inorganic and Organic Chemistry, Section of Organic Chemistry, Faculty of Chemistry, University of Barcelona, 08028 Barcelona, Spain
| | | | - Daniel Bexell
- Translational Cancer Research, Lund University, 22381 Lund, Sweden
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain, and CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Roland Nilsson
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
- Division of Cardiovascular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Jonas Bergquist
- Analytical Chemistry, Department of Chemistry and Science for Life Laboratory, Uppsala University, 751 24 Uppsala, Sweden
| | - Almut Schulze
- Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, 171 65 Stockholm, Sweden
| |
Collapse
|
9
|
Rae C, Fragkoulis GI, Chalmers AJ. Cytotoxicity and Radiosensitizing Activity of the Fatty Acid Synthase Inhibitor C75 Is Enhanced by Blocking Fatty Acid Uptake in Prostate Cancer Cells. Adv Radiat Oncol 2020; 5:994-1005. [PMID: 33083663 PMCID: PMC7557210 DOI: 10.1016/j.adro.2020.06.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/24/2020] [Accepted: 06/21/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancers, like many other types of cancer, express elevated levels of fatty acid synthase (FASN) to make more fatty acids, which are required for energy, signaling, and proliferation. Because inhibition of FASN has been shown to sensitize tumors to chemotherapy and radiation, we studied the effect of C75, a radiosensitizing FASN inhibitor, and compared its single agent and radiosensitizing activities in 2 prostate cancer cell lines, PC3 and LNCaP, with alternative FASN inhibitors that have progressed into clinical trials. We also investigated the effect of serum and fatty acid supplementation on responses to FASN inhibitors, probing expression of key proteins related to fatty acid uptake in response to FASN inhibition, irradiation, and serum lipid concentration and how this may be modulated to increase the potency of C75. We demonstrated that C75 was the only FASN inhibitor to sensitize cells to ionizing radiation; no sensitization was apparent with FASN inhibitors TVB-3166 or Orlistat. The prostate cancer cell lines were able to take up fatty acids from the culture medium, and the availability of fatty acids affected sensitivity of these cells to C75 but not the other FASN inhibitors tested. C75 also increased expression of fatty acid transporter proteins FATP1 and CD36. Furthermore, blocking CD36 with antibody increased the sensitivity of cells to C75. We suggest that the potency of C75 is affected by fatty acid availability and that the effectiveness of FASN inhibitors in combination with ionizing radiation can be further enhanced by regulating fatty acid uptake.
Collapse
Affiliation(s)
- Colin Rae
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Georgios I. Fragkoulis
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Anthony J. Chalmers
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
10
|
Tamarindo GH, Góes RM. Docosahexaenoic acid differentially modulates the cell cycle and metabolism- related genes in tumor and pre-malignant prostate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158766. [PMID: 32712248 DOI: 10.1016/j.bbalip.2020.158766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) has different molecular features along progression, including androgen profile, which is associated to therapy inefficiency leading to more aggressive phenotype. Docosahexaenoic acid (DHA) has antiproliferative and pro-apoptotic properties in different cancers associated to cell metabolism modulation. The latter is of particular interest since metabolic reprogramming is one of PCa hallmarks, but is not clear how this occurs among disease progression. Therefore, we evaluated DHA antiproliferative potential in distinct androgenic backgrounds associated to metabolism modulation and androgen-regulated genes. For this purpose, pre-malignant PNT1A and tumor AR-positive 22rv1, and AR-negative PC3 cells were incubated with DHA at 100 μM-48 h. DHA reduced at least 26% cell number for all lineages due to S-phase decrease in AR-positive and G2/M arrest in AR-negative. Mitochondrial metabolic rate decreased in PNT1A (~38%) and increased in tumor cells (at least 40%). This was associated with ROS overproduction (1.6-fold PNT1A; 2.1 22rv1; 2.2 PC3), lipid accumulation (3-fold PNT1A; 1.8 22rv1; 3.6 PC3) and mitochondria damage in all cell lines. AKT, AMPK and PTEN were not activated in any cell line, but p-ERK1/2 increased (1.5-fold) in PNT1A. Expression of androgen-regulated and nuclear receptors genes showed that DHA affected them in a distinct pattern in each cell line, but most converged to metabolism regulation, response to hormones, lipids and stress. In conclusion, regardless of androgenic or PTEN background DHA exerted antiproliferative effect associated to cell cycle impairment, lipid deregulation and oxidative stress, but differentially regulated gene expression probably due to distinct molecular features of each pathologic stage.
Collapse
Affiliation(s)
| | - Rejane Maira Góes
- Institute of Biology, University of Campinas, Campinas, SP, Brazil; Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, SP, Brazil.
| |
Collapse
|
11
|
Saleh A, ElFayoumi HM, Youns M, Barakat W. Rutin and orlistat produce antitumor effects via antioxidant and apoptotic actions. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:165-175. [PMID: 30465055 DOI: 10.1007/s00210-018-1579-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 11/05/2018] [Indexed: 12/17/2022]
Abstract
Cancer is a broad term used to describe a large number of diseases characterized by uncontrolled cell proliferation that leads to tumor production. Cancer is associated with mutations in genes controlling proliferation and apoptosis, oxidative stress, fatty acid synthase (FAS) expression, and other mechanisms. Currently, most antineoplastic drugs have severe adverse effects and new effective and safe drugs are needed. This study aims to investigate the possible anticancer activity of rutin and orlistat which are both safely used clinically in humans against two breast cancer models (in vivo EAC and in vitro MCF7) and the pancreatic cancer cell line (PANC-1). Our results have shown that both rutin and orlistat exerted an in vivo anticancer activity as evidenced by the decrease in tumor volume, CEA level, cholesterol content, FAS, and the exerted antioxidant action (reduced MDA level and increased GSH content) and through histopathological examination. In addition, both were cytotoxic to MCF-7 and Panc-1 cell lines by promoting apoptosis. In conclusion, the anticancer activity of rutin and orlistat makes them promising candidates for cancer treatment alone or in combination with other anticancer drugs specially that they are used clinically with an acceptable safety profile.
Collapse
Affiliation(s)
- Amira Saleh
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hassan M ElFayoumi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.,Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, Ismailia, Egypt
| | - Mahmoud Youns
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Helwan, Egypt.,Department of Biochemistry, Oman Pharmacy Institute, Ministry of Health, Muscat, Oman
| | - Waleed Barakat
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt. .,Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tabuk University, Tabuk, Kingdom of Saudi Arabia.
| |
Collapse
|
12
|
Rae C, Babich JW, Mairs RJ. Differential in radiosensitizing potency of enantiomers of the fatty acid synthase inhibitor C75. Chirality 2016; 29:10-13. [PMID: 27901292 PMCID: PMC5248592 DOI: 10.1002/chir.22668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 12/24/2022]
Abstract
The elevated activity of fatty acid synthase has been reported in a number of cancer types. Inhibition of this enzyme has been demonstrated to induce cancer cell death and reduce tumor growth. In addition, the fatty acid synthase inhibitor drug C75 has been reported to synergistically enhance the cancer‐killing ability of ionizing radiation. However, clinical use of C75 has been limited due to its producing weight loss, believed to be caused by alterations in the activity of carnitine palmitoyltransferase‐1. C75 is administered in the form of a racemic mixture of (−) and (+) enantiomers that may differ in their regulation of fatty acid synthase and carnitine palmitoyltransferase‐1. Therefore, we assessed the relative cancer‐killing potency of different enantiomeric forms of C75 in prostate cancer cells. These results suggest that (−)‐C75 is the more cytotoxic enantiomer and has greater radiosensitizing capacity than (+)‐C75. These observations will stimulate the development of fatty acid synthase inhibitors that are selective for cancer cells and enhance the tumor‐killing activity of ionizing radiation, while minimizing weight loss in cancer patients.
Collapse
Affiliation(s)
- Colin Rae
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - John W Babich
- Department of Radiology, Cornell University, Ithaca, New York, USA
| | - Robert J Mairs
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| |
Collapse
|
13
|
Zhang Z, Garzotto M, Beer TM, Thuillier P, Lieberman S, Mori M, Stoller WA, Farris PE, Shannon J. Effects of ω-3 Fatty Acids and Catechins on Fatty Acid Synthase in the Prostate: A Randomized Controlled Trial. Nutr Cancer 2016; 68:1309-1319. [PMID: 27646578 DOI: 10.1080/01635581.2016.1224365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal and human studies suggest fish oil and green tea may have protective effect on prostate cancer. Fatty acid synthase (FAS) has been hypothesized to be linked to chemoprotective effects of both compounds. This study evaluated the independent and joint effects of fish oil (FO) and green tea supplement (epigallocatechin-3-gallate, EGCG) on FAS and Ki-67 levels in prostate tissue. Through a double-blinded, randomized controlled trial with 2 × 2 factorial design, 89 men scheduled for repeat prostate biopsy following an initial negative prostate biopsy were randomized into either FO alone (1.9 g DHA + EPA/day), EGCG alone (600 mg/day), a combination of FO and EGCG, or placebo. We used linear mixed-effects models to test the differences of prostate tissue FAS and Ki-67 by immunohistochemistry between pre- and post-intervention within each group, as well as between treatment groups. Results did not show significant difference among treatment groups in pre-to-post-intervention changes of FAS (P = 0.69) or Ki-67 (P = 0.26). Comparing placebo group with any of the treatment groups, we did not find significant difference in FAS or Ki-67 changes (all P > 0.05). Results indicate FO or EGCG supplementation for a short duration may not be sufficient to produce biologically meaningful changes in FAS or Ki-67 levels in prostate tissue.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA
| | - Mark Garzotto
- b Department of Urology , Portland Veterans Administration Medical Center, Oregon Health and Science University , Portland , Oregon , USA
| | - Tomasz M Beer
- c Knight Cancer Institute, Oregon Health and Science University , Portland , Oregon , USA
| | - Philippe Thuillier
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA.,c Knight Cancer Institute, Oregon Health and Science University , Portland , Oregon , USA.,d Department of Dermatology , Oregon Health and Science University , Portland , Oregon , USA
| | - Stephen Lieberman
- e Department of Urology , Kaiser Permanente Northwest , Clackamas , Oregon , USA
| | - Motomi Mori
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA.,c Knight Cancer Institute, Oregon Health and Science University , Portland , Oregon , USA
| | - Wesley A Stoller
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA
| | - Paige E Farris
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA
| | - Jackilen Shannon
- a OHSU-PSU School of Public Health, Oregon Health and Science University , Portland , Oregon , USA
| |
Collapse
|
14
|
Wen S, Tian J, Niu Y, Li L, Yeh S, Chang C. ASC-J9®, and not Casodex or Enzalutamide, suppresses prostate cancer stem/progenitor cell invasion via altering the EZH2-STAT3 signals. Cancer Lett 2016; 376:377-86. [DOI: 10.1016/j.canlet.2016.01.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/03/2015] [Accepted: 01/11/2016] [Indexed: 11/27/2022]
|
15
|
Wen S, Niu Y, Lee SO, Yeh S, Shang Z, Gao H, Li Y, Chou F, Chang C. Targeting fatty acid synthase with ASC-J9 suppresses proliferation and invasion of prostate cancer cells. Mol Carcinog 2016; 55:2278-2290. [PMID: 26894509 DOI: 10.1002/mc.22468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 01/09/2016] [Accepted: 01/15/2016] [Indexed: 01/22/2023]
Abstract
Fatty acid synthase (FASN) is the key enzyme for the control of fatty acid synthesis that contributes significantly to the prostate cancer (PCa) progression. It was reported that androgens were able to induce FASN expression in PCa, and addition of the anti-androgen Casodex might suppress the androgen-induced FASN expression. However, here we found androgen-deprivation-therapy (ADT) with anti-androgens Bicalutamide (Casodex) or Enzalutamide (MDV3100) had little effect to suppress FASN expression and FASN-mediated cell growth and invasion during the castration resistant stage when the androgen concentration is 1 nM DHT (dihydrotestosterone). In contrast, the newly developed androgen receptor (AR) degradation enhancer ASC-J9® suppressed FASN expression and FASN-mediated cell growth and invasion in various PCa cell lines at 1 nM DHT. Mechanism dissection found ASC-J9® could suppress significantly the FASN expression and FASN-mediated PCa progression via the AR-dependent pathway involving AR→SREBP-1→FASN signaling in AR-positive C4-2 and LNCaP cells and via the AR-independent pathway involving the modulation of PI3K/AKT→SREBP-1→FASN signaling in AR-negative PC-3 and DU145 cells. Together, these results suggest that FASN is one of the important mechanism why the current ADT eventually fails. ASC-J9® might represent a new potential therapeutic approach to suppress FASN-mediated PCa progression via both AR-dependent and AR-independent pathways during the castration resistant stage of PCa. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simeng Wen
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China.,Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Soo Ok Lee
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Shuyuan Yeh
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Zhiqun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Hengheng Gao
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Yanjun Li
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, University of Tianjin Medical, Tianjin, China
| | - Fuju Chou
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, Rochester, New York.,Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan
| |
Collapse
|
16
|
Zhao H, Pflug BR, Lai X, Wang M. Metabolic and molecular regulation of dietary polyunsaturated fatty acids on prostate cancer. Proteomics Clin Appl 2015; 10:267-79. [DOI: 10.1002/prca.201500066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/07/2015] [Accepted: 11/11/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Heng Zhao
- Department of Biochemistry and Molecular Biology; Indiana University School of Medicine; Indianapolis IN USA
| | - Beth R. Pflug
- Department of Medicine, Division of Clinical Pharmacology; Indiana University School of Medicine; Indianapolis IN USA
| | - Xianyin Lai
- Department of Biochemistry and Molecular Biology; Indiana University School of Medicine; Indianapolis IN USA
- Department of Cellular and Integrative Physiology; Indiana University School of Medicine; Indianapolis IN USA
| | - Mu Wang
- Department of Biochemistry and Molecular Biology; Indiana University School of Medicine; Indianapolis IN USA
| |
Collapse
|
17
|
Rae C, Haberkorn U, Babich JW, Mairs RJ. Inhibition of Fatty Acid Synthase Sensitizes Prostate Cancer Cells to Radiotherapy. Radiat Res 2015; 184:482-93. [DOI: 10.1667/rr14173.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
18
|
Ritterson Lew C, Guin S, Theodorescu D. Targeting glycogen metabolism in bladder cancer. Nat Rev Urol 2015; 12:383-91. [PMID: 26032551 DOI: 10.1038/nrurol.2015.111] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metabolism has been a heavily investigated topic in cancer research for the past decade. Although the role of aerobic glycolysis (the Warburg effect) in cancer has been extensively studied, abnormalities in other metabolic pathways are only just being understood in cancer. One such pathway is glycogen metabolism; its involvement in cancer development, particularly in urothelial malignancies, and possible ways of exploiting aberrations in this process for treatment are currently being studied. New research shows that the glycogen debranching enzyme amylo-α-1,6-glucosidase, 4-α-glucanotransferase (AGL) is a novel tumour suppressor in bladder cancer. Loss of AGL leads to rapid proliferation of bladder cancer cells. Another enzyme involved in glycogen debranching, glycogen phosphorylase, has been shown to be a tumour promoter in cancer, including in prostate cancer. Studies demonstrate that bladder cancer cells in which AGL expression is lost are more metabolically active than cells with intact AGL expression, and these cells are more sensitive to inhibition of both glycolysis and glycine synthesis--two targetable pathways. As a tumour promoter and enzyme, glycogen phosphorylase can be directly targeted, and preclinical inhibitor studies are promising. However, few of these glycogen phosphorylase inhibitors have been tested for cancer treatment in the clinical setting. Several possible limitations to the targeting of AGL and glycogen phosphorylase might also exist.
Collapse
Affiliation(s)
- Carolyn Ritterson Lew
- Department of Surgery (Urology), University of Colorado, 12700 East 19th Avenue, RC2/P15-6430D/MS-8609, Aurora, CO 80045, USA
| | - Sunny Guin
- Department of Surgery (Urology), University of Colorado, 12700 East 19th Avenue, RC2/P15-6430D/MS-8609, Aurora, CO 80045, USA
| | - Dan Theodorescu
- University of Colorado Comprehensive Cancer Center, MS F-434, 13001 East 17th Place, Aurora, CO 80045, USA
| |
Collapse
|
19
|
Bauerschlag DO, Maass N, Leonhardt P, Verburg FA, Pecks U, Zeppernick F, Morgenroth A, Mottaghy FM, Tolba R, Meinhold-Heerlein I, Bräutigam K. Fatty acid synthase overexpression: target for therapy and reversal of chemoresistance in ovarian cancer. J Transl Med 2015; 13:146. [PMID: 25947066 PMCID: PMC4504229 DOI: 10.1186/s12967-015-0511-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/29/2015] [Indexed: 12/30/2022] Open
Abstract
Background Fatty acid synthase (FASN) is crucial to de novo long-chain fatty acid synthesis, needed to meet cancer cells’ increased demands for membrane, energy, and protein production. Methods We investigated FASN overexpression as a therapeutic and chemosensitization target in ovarian cancer tissue, cell lines, and primary cell cultures. FASN expression at mRNA and protein levels was determined by quantitative real-time polymerase chain reaction and immunoblotting and immunohistochemistry, respectively. FASN inhibition’s impact on cell viability, apoptosis, and fatty acid metabolism was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium-bromide assay, cell death detection enzyme-linked immunosorbent assay, immunoblotting, and 18 F-fluoromethylcholine uptake measurement, respectively. Results Relative to that in healthy fallopian tube tissue, tumor tissues had 1.8-fold average FASN protein overexpression; cell lines and primary cultures had 11-fold–100-fold mRNA and protein overexpression. In most samples, the FASN inhibitor cerulenin markedly decreased FASN expression and cell viability and induced apoptosis. Unlike concomitant administration, sequential cerulenin/cisplatin treatment reduced cisplatin’s half maximal inhibitory concentration profoundly (up to 54%) in a cisplatin-resistant cell line, suggesting platinum (re)sensitization. Cisplatin-resistant cells displayed lower 18 F-fluoro-methylcholine uptake than did cisplatin-sensitive cells, suggesting that metabolic imaging might help guide therapy. Conclusions FASN inhibition induced apoptosis in chemosensitive and platinum-resistant ovarian cancer cells and may reverse cisplatin resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0511-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dirk O Bauerschlag
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Peter Leonhardt
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Frederik A Verburg
- Department of Nuclear Medicine, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Ulrich Pecks
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Felix Zeppernick
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Rene Tolba
- Institute for Laboratory Animal Science, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Ivo Meinhold-Heerlein
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Karen Bräutigam
- Department of Gynecology and Obstetrics, University Medical Center RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
20
|
Salicylate activates AMPK and synergizes with metformin to reduce the survival of prostate and lung cancer cells ex vivo through inhibition of de novo lipogenesis. Biochem J 2015; 469:177-87. [PMID: 25940306 DOI: 10.1042/bj20150122] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/05/2015] [Indexed: 01/04/2023]
Abstract
Aspirin, the pro-drug of salicylate, is associated with reduced incidence of death from cancers of the colon, lung and prostate and is commonly prescribed in combination with metformin in individuals with type 2 diabetes. Salicylate activates the AMP-activated protein kinase (AMPK) by binding at the A-769662 drug binding site on the AMPK β1-subunit, a mechanism that is distinct from metformin which disrupts the adenylate charge of the cell. A hallmark of many cancers is high rates of fatty acid synthesis and AMPK inhibits this pathway through phosphorylation of acetyl-CoA carboxylase (ACC). It is currently unknown whether targeting the AMPK-ACC-lipogenic pathway using salicylate and/or metformin may be effective for inhibiting cancer cell survival. Salicylate suppresses clonogenic survival of prostate and lung cancer cells at therapeutic concentrations achievable following the ingestion of aspirin (<1.0 mM); effects not observed in prostate (PNT1A) and lung (MRC-5) epithelial cell lines. Salicylate concentrations of 1 mM increased the phosphorylation of ACC and suppressed de novo lipogenesis and these effects were enhanced with the addition of clinical concentrations of metformin (100 μM) and eliminated in mouse embryonic fibroblasts (MEFs) deficient in AMPK β1. Supplementation of media with fatty acids and/or cholesterol reverses the suppressive effects of salicylate and metformin on cell survival indicating the inhibition of de novo lipogenesis is probably important. Pre-clinical studies evaluating the use of salicylate based drugs alone and in combination with metformin to inhibit de novo lipogenesis and the survival of prostate and lung cancers are warranted.
Collapse
|
21
|
Cheng J, Ondracek RP, Mehedint DC, Kasza KA, Xu B, Gill S, Azabdaftari G, Yao S, Morrison CD, Mohler JL, Marshall JR. Association of fatty-acid synthase polymorphisms and expression with outcomes after radical prostatectomy. Prostate Cancer Prostatic Dis 2015; 18:182-9. [PMID: 25868764 DOI: 10.1038/pcan.2015.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/24/2015] [Accepted: 02/28/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Fatty-acid synthase (FASN), selectively overexpressed in prostate cancer (PCa) cells, has been described as linked to the aggressiveness of PCa. Constitutional genetic variation of the FASN gene and the expression levels of FASN protein in cancer cells could thus be expected to predict outcome after radical prostatectomy (RP). This study evaluates the associations of malignant tissue status, neoadjuvant androgen deprivation therapy (NADT) and single-nucleotide polymorphisms (SNPs) of FASN with FASN protein expression in prostate tissue. The study then examines the associations of FASN SNPs and gene expression with three measures of post-prostatectomy outcome. METHODS Seven tagging FASN SNPs were genotyped in 659 European American men who underwent RP at Roswell Park Cancer Institute between 1993 and 2005. FASN protein expression was assessed using immunohistochemistry. The patients were followed for an average of 6.9 years (range: 0.1-20.6 years). Outcome was assessed using three end points: biochemical failure, treatment failure and development of distant metastatic PCa. Cox proportional hazards analyses were used to evaluate the associations of the tagging SNPs and FASN expression with these end points. Bivariate associations with outcomes were considered; the associations also were controlled for known aggressiveness indicators. RESULTS Overall, no SNPs were associated with any known aggressiveness indicators. FASN staining intensity was stronger in malignant than in benign tissue, and NADT was associated with decreased FASN staining in both benign and malignant tissue. The relationships of FASN SNPs and staining intensity with outcome were less clear. One SNP, rs4246444, showed a weak association with outcome. FASN staining intensity also showed a weak and seemingly contradictory relationship with outcome. CONCLUSIONS Additional study with longer follow-up and populations that include more metastatic patients is warranted.
Collapse
Affiliation(s)
- J Cheng
- 1] Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA [2] Department of Pathology, University at Buffalo, Buffalo, NY, USA
| | - R P Ondracek
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - D C Mehedint
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K A Kasza
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - B Xu
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Gill
- Department of Pathology, University at Buffalo, Buffalo, NY, USA
| | - G Azabdaftari
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - S Yao
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - C D Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - J L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - J R Marshall
- Department of Cancer Prevention and Population Science, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
22
|
Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, Goodwin LM, Smethurst E, Mason S, Blyth K, McGarry L, James D, Shanks E, Kalna G, Saunders RE, Jiang M, Howell M, Lassailly F, Thin MZ, Spencer-Dene B, Stamp G, van den Broek NJF, Mackay G, Bulusu V, Kamphorst JJ, Tardito S, Strachan D, Harris AL, Aboagye EO, Critchlow SE, Wakelam MJO, Schulze A, Gottlieb E. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 2015; 27:57-71. [PMID: 25584894 PMCID: PMC4297291 DOI: 10.1016/j.ccell.2014.12.002] [Citation(s) in RCA: 553] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 12/17/2022]
Abstract
A functional genomics study revealed that the activity of acetyl-CoA synthetase 2 (ACSS2) contributes to cancer cell growth under low-oxygen and lipid-depleted conditions. Comparative metabolomics and lipidomics demonstrated that acetate is used as a nutritional source by cancer cells in an ACSS2-dependent manner, and supplied a significant fraction of the carbon within the fatty acid and phospholipid pools. ACSS2 expression is upregulated under metabolically stressed conditions and ACSS2 silencing reduced the growth of tumor xenografts. ACSS2 exhibits copy-number gain in human breast tumors, and ACSS2 expression correlates with disease progression. These results signify a critical role for acetate consumption in the production of lipid biomass within the harsh tumor microenvironment.
Collapse
Affiliation(s)
- Zachary T Schug
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Barrie Peck
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Dylan T Jones
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Qifeng Zhang
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Israt S Alam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | | | - Susan Mason
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Blyth
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Lynn McGarry
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Daniel James
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Emma Shanks
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Gabriela Kalna
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Rebecca E Saunders
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Ming Jiang
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Michael Howell
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Francois Lassailly
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - May Zaw Thin
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Bradley Spencer-Dene
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Gordon Stamp
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Niels J F van den Broek
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Gillian Mackay
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Vinay Bulusu
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jurre J Kamphorst
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Saverio Tardito
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - David Strachan
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | | | - Almut Schulze
- Cancer Research UK, London Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Eyal Gottlieb
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
23
|
Jurczyszyn A, Czepiel J, Gdula-Argasińska J, Paśko P, Czapkiewicz A, Librowski T, Perucki W, Butrym A, Castillo JJ, Skotnicki AB. Plasma fatty acid profile in multiple myeloma patients. Leuk Res 2014; 39:400-5. [PMID: 25666255 DOI: 10.1016/j.leukres.2014.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/01/2014] [Accepted: 12/16/2014] [Indexed: 11/30/2022]
Abstract
New membrane formation in the proliferating tumor cells consequently results in hypermetabolism of fatty acids (FA), as seen in many cancer patients, including multiple myeloma (MM). The FA composition of plasma reflects both endogenous synthesis as well as the dietary supply of these compounds. Additionally, obesity is a risk factor for the development of MM. The aim of this study was to compare the FA composition of plasma in 60 MM patients and 60 healthy controls. We noted significant differences in the FA profile of plasma from patients with MM when compared to the control group. Increased levels of saturated and n-6 polyunsaturated fatty acids in MM patients suggest that there may be increased endogenous synthesis of these fatty acids, likely due to increased expression of desaturase and elongase. Furthermore, cluster analysis showed differences in the distribution of FA in plasma from MM patients compared to controls. Dietary fat and a deranged endogenous FA metabolism may contribute to cancer-associated inflammation through an abnormal arachidonic acid metabolism, caused by pro-inflammatory derivatives. Our study supports further research on the biochemistry of lipids in patients with MM.
Collapse
Affiliation(s)
| | - Jacek Czepiel
- Department of Infectious Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Gdula-Argasińska
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Paweł Paśko
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Czapkiewicz
- Faculty of Management, AGH University of Science and Technology, Krakow, Poland
| | - Tadeusz Librowski
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - William Perucki
- Department of Medicine, John Dempsey Hospital, University of Connecticut, Farmington, CT, USA
| | - Aleksandra Butrym
- Department of Haematology, Blood Neoplasms and Bone Marrow Transplantation, Medical University, Wroclaw, Poland; Department of Physiology, Medical University, Wroclaw, Poland
| | - Jorge J Castillo
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
24
|
Jurczyszyn A, Czepiel J, Gdula-Argasińska J, Czapkiewicz A, Biesiada G, Dróżdż M, Perucki W, Castillo JJ. Erythrocyte membrane fatty acids in multiple myeloma patients. Leuk Res 2014; 38:1260-5. [PMID: 25192858 DOI: 10.1016/j.leukres.2014.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 07/09/2014] [Accepted: 08/16/2014] [Indexed: 01/01/2023]
Abstract
Mounting data show that fatty acids (FA) and fatty acid synthase (FAS) function could be potential targets for multiple myeloma (MM) therapy. Our study aimed at comparing the FA composition of erythrocyte membranes of MM patients and healthy controls. MM patients had higher saturated FA and n-6 polyunsaturated FA (PUFA) and lower monounsaturated, n-3 PUFA and trans-FA indices than controls. The n-3/n-6 PUFA ratio was lower in MM patients and there was distinct clustering of variants of individual FA in MM patients. The FA content of erythrocyte membrane could serve as a diagnostic and/or predictive biomarker in MM.
Collapse
Affiliation(s)
| | - Jacek Czepiel
- Department of Infectious Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Gdula-Argasińska
- Department of Radioligands, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Czapkiewicz
- Faculty of Management, AGH University of Science and Technology, Krakow, Poland
| | - Grażyna Biesiada
- Department of Infectious Diseases, Jagiellonian University Medical College, Krakow, Poland
| | | | - William Perucki
- Students' Scientific Society, Jagiellonian University Medical College, Krakow, Poland
| | - Jorge J Castillo
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
25
|
Madigan AA, Rycyna KJ, Parwani AV, Datiri YJ, Basudan AM, Sobek KM, Cummings JL, Basse PH, Bacich DJ, O'Keefe DS. Novel nuclear localization of fatty acid synthase correlates with prostate cancer aggressiveness. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2156-62. [PMID: 24907642 DOI: 10.1016/j.ajpath.2014.04.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 04/16/2014] [Accepted: 04/28/2014] [Indexed: 12/24/2022]
Abstract
Fatty acid synthase is up-regulated in a variety of cancers, including prostate cancer. Up-regulation of fatty acid synthase not only increases production of fatty acids in tumors but also contributes to the transformed phenotype by conferring growth and survival advantages. In addition, increased fatty acid synthase expression in prostate cancer correlates with poor prognosis, although the mechanism(s) by which this occurs are not completely understood. Because fatty acid synthase is expressed at low levels in normal cells, it is currently a major target for anticancer drug design. Fatty acid synthase is normally found in the cytosol; however, we have discovered that it also localizes to the nucleus in a subset of prostate cancer cells. Analysis of the fatty acid synthase protein sequence indicated the presence of a nuclear localization signal, and subcellular fractionation of LNCaP prostate cancer cells, as well as immunofluorescent confocal microscopy of patient prostate tumor tissue and LNCaPs confirmed nuclear localization of this protein. Finally, immunohistochemical analysis of prostate cancer tissue indicated that nuclear localization of fatty acid synthase correlates with Gleason grade, implicating a potentially novel role in prostate cancer progression. Possible clinical implications include improving the accuracy of prostate biopsies in the diagnosis of low- versus intermediate-risk prostate cancer and the uncovering of novel metabolic pathways for the therapeutic targeting of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Allison A Madigan
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kevin J Rycyna
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Ahmed M Basudan
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kathryn M Sobek
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica L Cummings
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Per H Basse
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Dean J Bacich
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania; University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Denise S O'Keefe
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania; University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
26
|
Hamada S, Horiguchi A, Asano T, Kuroda K, Asakuma J, Ito K, Asano T, Miyai K, Iwaya K. Prognostic impact of fatty acid synthase expression in upper urinary tract urothelial carcinoma. Jpn J Clin Oncol 2014; 44:486-92. [PMID: 24643292 DOI: 10.1093/jjco/hyu020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Fatty acid synthase has been shown to be highly expressed in various types of cancers with increased tumour aggressiveness. In this study we examined the level of fatty acid synthase expression in surgically resected upper urinary tract urothelial carcinoma specimens and evaluated the relations between fatty acid synthase expression and the patients' pathological features and clinical outcomes. METHODS Sections of paraffin-embedded tumour specimens from 113 patients who underwent surgical treatment for upper urinary tract urothelial carcinoma were immunostained with a polyclonal fatty acid synthase antibody, and a tumour was considered to have high fatty acid synthase expression if >50% of the cancer cells stained with moderate-to-strong intensity. Associations between fatty acid synthase expression and the patients' pathological parameters and survival were analyzed statistically. RESULTS During the follow-up time (median: 46.8 months), 61 patients (54.0%) had recurrence and 17 (15.0%) died of upper urinary tract urothelial carcinoma. High fatty acid synthase expression was significantly associated with high tumour grade (P = 0.0273). Patients with high fatty acid synthase expression had significantly worse recurrence-free survival and extravesical-recurrence-free survival than those with low fatty acid synthase expression (P = 0.0171, P = 0.0228, respectively). In multivariate analysis, high fatty acid synthase expression was an independent predictor of shortened recurrence-free survival (P = 0.0220, hazard ratio (HR) = 1.970). CONCLUSIONS Fatty acid synthase expression in upper urinary tract urothelial carcinoma is an independent predictor for tumour recurrence. Patients with high fatty acid synthase expression in upper urinary tract urothelial carcinoma should be followed carefully and adjuvant therapy for them should be considered.
Collapse
Affiliation(s)
- Shinsuke Hamada
- *Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Swierczynski J, Hebanowska A, Sledzinski T. Role of abnormal lipid metabolism in development, progression, diagnosis and therapy of pancreatic cancer. World J Gastroenterol 2014; 20:2279-303. [PMID: 24605027 PMCID: PMC3942833 DOI: 10.3748/wjg.v20.i9.2279] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/25/2013] [Accepted: 01/03/2014] [Indexed: 02/07/2023] Open
Abstract
There is growing evidence that metabolic alterations play an important role in cancer development and progression. The metabolism of cancer cells is reprogrammed in order to support their rapid proliferation. Elevated fatty acid synthesis is one of the most important aberrations of cancer cell metabolism. An enhancement of fatty acids synthesis is required both for carcinogenesis and cancer cell survival, as inhibition of key lipogenic enzymes slows down the growth of tumor cells and impairs their survival. Based on the data that serum fatty acid synthase (FASN), also known as oncoantigen 519, is elevated in patients with certain types of cancer, its serum level was proposed as a marker of neoplasia. This review aims to demonstrate the changes in lipid metabolism and other metabolic processes associated with lipid metabolism in pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic neoplasm, characterized by high mortality. We also addressed the influence of some oncogenic factors and tumor suppressors on pancreatic cancer cell metabolism. Additionally the review discusses the potential role of elevated lipid synthesis in diagnosis and treatment of pancreatic cancer. In particular, FASN is a viable candidate for indicator of pathologic state, marker of neoplasia, as well as, pharmacological treatment target in pancreatic cancer. Recent research showed that, in addition to lipogenesis, certain cancer cells can use fatty acids from circulation, derived from diet (chylomicrons), synthesized in liver, or released from adipose tissue for their growth. Thus, the interactions between de novo lipogenesis and uptake of fatty acids from circulation by PDAC cells require further investigation.
Collapse
|
28
|
Hamada S, Horiguchi A, Kuroda K, Ito K, Asano T, Miyai K, Iwaya K. Increased fatty acid synthase expression in prostate biopsy cores predicts higher Gleason score in radical prostatectomy specimen. BMC Clin Pathol 2014; 14:3. [PMID: 24422874 PMCID: PMC3898069 DOI: 10.1186/1472-6890-14-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 01/09/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fatty acid synthase (FAS) is highly expressed in various types of cancer, and elevated expression of FAS has been suggested to be a predictor of tumor aggressiveness and poor prognosis. We examined whether FAS expression in prostate biopsy cores could predict the pathological characteristics of radical prostatectomy (RP) specimens. METHODS Paraffin-embedded prostate biopsy cores, obtained from 102 patients who subsequently underwent RP, were immunostained with polyclonal anti-FAS antibody. The staining intensity was categorized into non-staining, weak, moderate, and strong. Tumors with moderate or strong immunostaining were considered to show high FAS expression, and other tumors were considered to show low FAS expression. The relation between the FAS expression status in biopsy cores and pathological parameters in RP specimens was analyzed. RESULTS The FAS expression in the biopsy cores of 64 of the 102 tumors (63%) was high, whereas it was low in the biopsy cores of the other 38 tumors (37%). High FAS expression was significantly associated with Gleason Score (GS) ≥ 7 in RP specimens (p< 0.0001). In multivariable logistic regression analyses, GS ≥7 in biopsy cores (p <0.0001), higher preoperative PSA (p = 0.0194), and high FAS expression (p = 0.0004) were independent predictors of GS ≥ 7 in the RP specimen. CONCLUSIONS Increased FAS expression in prostate biopsy cores could be a novel parameter for predicting higher GS in RP specimens. The treatment strategy for patients with high FAS expression in prostate biopsy cores should be carefully determined.
Collapse
Affiliation(s)
- Shinsuke Hamada
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa-City, Saitama 359-8513, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Ito T, Sato K, Maekawa H, Sakurada M, Orita H, Shimada K, Daida H, Wada R, Abe M, Hino O, Kajiyama Y. Elevated levels of serum fatty acid synthase in patients with gastric carcinoma. Oncol Lett 2014; 7:616-620. [PMID: 24527066 PMCID: PMC3919915 DOI: 10.3892/ol.2014.1793] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 12/06/2013] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the second leading cause of cancer mortality in the world. It is important to develop biomarkers for detecting new cancers at an early stage and for treating them early during recurrence in order to guide optimal treatment. Fatty acid synthase (FAS) is highly expressed in numerous human cancers and thus could potentially serve as such a biomarker, but the potential utility of measuring FAS for detecting gastric cancer has not been previously investigated. The aim of the present study was to provide a preliminary assessment of serum FAS as a marker of gastric carcinoma. The study included 47 patients with gastric cancer and 150 healthy subjects. Blood samples were collected from each cancer patient prior to treatment. Serum FAS levels were measured by ELISA and compared across the two groups of patients. Significantly higher levels of serum FAS were found in the gastric cancer patients [95% confidence interval (CI), 30.37–52.46] compared with the healthy controls (95% CI, 1.331–2.131), with elevated levels even in patients with early-stage tumors. These results indicate that measuring serum FAS levels has strong potential to provide a biomarker for the detection of gastric cancer, with high sensitivity and specificity.
Collapse
Affiliation(s)
- Tomoaki Ito
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Koichi Sato
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Hiroshi Maekawa
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Mutsumi Sakurada
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Hajime Orita
- Department of Surgery, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Kazunori Shimada
- Department of Cardiovascular Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryo Wada
- Department of Pathology, Juntendo Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Masaaki Abe
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Okio Hino
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiaki Kajiyama
- Department of Esophageal and Gastroenterological Surgery, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Fatty acid synthase overexpression in adult testicular germ cell tumors: potential role in the progression of non-seminomatous germ cell tumors. Virchows Arch 2013; 464:221-8. [PMID: 24337182 DOI: 10.1007/s00428-013-1525-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/09/2013] [Accepted: 12/03/2013] [Indexed: 10/25/2022]
Abstract
Overexpression of fatty acid synthase (FASN), which is a key enzyme responsible for the endogenous synthesis of fatty acids, and its association with multistep progression have been demonstrated in various human malignant tumors. We aimed to clarify the potential role of FASN overexpression in the development and progression of adult testicular germ cell tumors (TGCTs). From the primary sites of a cohort of 113 TGCT cases, we obtained 221 histological components: 53 intratubular germ cell neoplasias, unclassified (IGCNUs), 84 seminomas, 32 embryonal carcinomas, seven choriocarcinomas, 21 yolk sac tumors, and 24 teratomas. Samples were analyzed for overexpression of FASN by immunohistochemistry. Intensities of immunoreactivity and the fraction of positive cells were classified into each four categories (intensity, 0 to 3; fraction, 0-10 % = 1, 11-50 % = 2, 51-80 % = 3, and >80 % = 4). The overall score was determined by multiplication of both scores and overall scores greater than 6 were considered FASN overexpression. On a component basis, FASN overexpression was detected in 8 % of seminomas but not in IGCNUs (0 %) and was detected frequently in non-seminomatous germ cell tumors (NSGCTs) (88 % of embryonal carcinomas, all choriocarcinomas, 81 % of yolk sac tumors, and 54 % of teratomas). There were no cases of a mixed tumor (i.e., a tumor with multiple histological components) that overexpressed FASN in seminoma components but not in co-existing NSGCT components, suggesting sequential progression. Our immunohistochemical data suggest that FASN overexpression occurs as a late event during the progression from IGCNUs/seminomas to NSGCTs.
Collapse
|
31
|
Ridgway ND. The role of phosphatidylcholine and choline metabolites to cell proliferation and survival. Crit Rev Biochem Mol Biol 2013; 48:20-38. [PMID: 23350810 DOI: 10.3109/10409238.2012.735643] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The reorganization of metabolic pathways in cancer facilitates the flux of carbon and reducing equivalents into anabolic pathways at the expense of oxidative phosphorylation. This provides rapidly dividing cells with the necessary precursors for membrane, protein and nucleic acid synthesis. A fundamental metabolic perturbation in cancer is the enhanced synthesis of fatty acids by channeling glucose and/or glutamine into cytosolic acetyl-CoA and upregulation of key biosynthetic genes. This lipogenic phenotype also extends to the production of complex lipids involved in membrane synthesis and lipid-based signaling. Cancer cells display sensitivity to ablation of fatty acid synthesis possibly as a result of diminished capacity to synthesize complex lipids involved in signaling or growth pathways. Evidence has accrued that phosphatidylcholine, the major phospholipid component of eukaryotic membranes, as well as choline metabolites derived from its synthesis and catabolism, contribute to both proliferative growth and programmed cell death. This review will detail our current understanding of how coordinated changes in substrate availability, gene expression and enzyme activity lead to altered phosphatidylcholine synthesis in cancer, and how these changes contribute directly or indirectly to malignant growth. Conversely, apoptosis targets key steps in phosphatidylcholine synthesis and degradation that are linked to disruption of cell cycle regulation, reinforcing the central role that phosphatidylcholine and its metabolites in determining cell fate.
Collapse
Affiliation(s)
- Neale D Ridgway
- Departments of Pediatrics and Biochemistry & Molecular Biology, The Atlantic Research Centre, Dalhousie University, Halifax, Nova Scotia Canada.
| |
Collapse
|
32
|
Clinical applications of recent molecular advances in urologic malignancies: no longer chasing a "mirage"? Adv Anat Pathol 2013; 20:175-203. [PMID: 23574774 DOI: 10.1097/pap.0b013e3182863f80] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As our understanding of the molecular events leading to the development and progression of genitourologic malignancies, new markers of detection, prognostication, and therapy prediction can be exploited in the management of these prevalent tumors. The current review discusses the recent advances in prostate, bladder, renal, and testicular neoplasms that are pertinent to the anatomic pathologist.
Collapse
|
33
|
Dufour J, Pommier A, Alves G, De Boussac H, Lours-Calet C, Volle DH, Lobaccaro JMA, Baron S. Lack of liver X receptors leads to cell proliferation in a model of mouse dorsal prostate epithelial cell. PLoS One 2013; 8:e58876. [PMID: 23554947 PMCID: PMC3595217 DOI: 10.1371/journal.pone.0058876] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/07/2013] [Indexed: 11/19/2022] Open
Abstract
Recent studies underline the implication of Liver X Receptors (LXRs) in several prostate diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. In order to understand the molecular mechanisms involved, we derived epithelial cells from dorsal prostate (MPECs) of wild type (WT) or Lxrαβ−/− mice. In the WT MPECs, our results show that LXR activation reduces proliferation and correlates with the modification of the AKT-survival pathway. Moreover, LXRs regulate lipid homeostasis with the regulation of Abca1, Abcg1 and Idol, and, in a lesser extent, Srebp1, Fas and Acc. Conversely cells derived from Lxrαβ−/− mice show a higher basal phosphorylation and consequently activation of the survival/proliferation transduction pathways AKT and MAPK. Altogether, our data point out that the cell model we developed allows deciphering the molecular mechanisms inducing the cell cycle arrest. Besides, we show that activated LXRs regulate AKT and MAPK transduction pathways and demonstrate that LXRs could be good pharmacological targets in prostate disease such as cancer.
Collapse
Affiliation(s)
- Julie Dufour
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Aurélien Pommier
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Georges Alves
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Hugues De Boussac
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Corinne Lours-Calet
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - David H. Volle
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Jean-Marc A. Lobaccaro
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
| | - Silvère Baron
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement (GReD), Clermont-Ferrand, France
- CNRS, UMR 6293, GReD, Aubiere, France
- INSERM, UMR 1103, GReD, Aubiere, France
- Centre de Recherche en Nutrition Humaine d’Auvergne, Clermont-Ferrand, France
- * E-mail:
| |
Collapse
|
34
|
Marine- and plant-derived ω-3 fatty acids differentially regulate prostate cancer cell proliferation. Mol Clin Oncol 2013; 1:444-452. [PMID: 24649190 DOI: 10.3892/mco.2013.76] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/05/2013] [Indexed: 12/30/2022] Open
Abstract
Fish oil contains the marine ω-3 polyunsaturated fatty acids (ω-3 PUFAs) docosahexaenoic (DHA) and eicosapentaenoic acid (EPA). The consumption of diets rich in these fatty acids is associated with a decreased incidence of prostate cancer. However, there is limited knowledge regarding the non-marine ω-3 PUFA α-linolenic acid (ALA). To study which ω-3 PUFAs are more effective in prostate cancer prevention, and whether the mechanisms of action are conserved between them, we investigated the effect of DHA, EPA and ALA on the human prostate cancer cell lines PC-3 and LNCaP. Different trends of inhibition of PC-3 cell proliferation were observed for the three ω-3 PUFA, with DHA having the most pronounced effects on cell proliferation, while ALA had the minimum effects of the three ω-3 PUFAs. All the ω-3 PUFAs decreased fatty acid synthase (FASN) mRNA. Concerning genes involved in inflammation, cell cycle and apoptosis, DHA regulated the most genes in all categories, followed by EPA and then ALA. In addition, DHA and EPA increased the gene expression of the pro-apoptotic protein activating transcription factor 3 mRNA. Moreover, these two fatty acids significantly induced apoptosis. In conclusion, while some mechanisms of cancer cell inhibition are conserved among ω-3 PUFA, the extent, magnitude, and duration of transcriptional changes vary for each individual fatty acid.
Collapse
|
35
|
Zu X, Zhong J, Luo D, Tan J, Zhang Q, Wu Y, Liu J, Cao R, Wen G, Cao D. Chemical genetics of acetyl-CoA carboxylases. Molecules 2013; 18:1704-19. [PMID: 23358327 PMCID: PMC6269866 DOI: 10.3390/molecules18021704] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 01/03/2013] [Accepted: 01/11/2013] [Indexed: 12/16/2022] Open
Abstract
Chemical genetic studies on acetyl-CoA carboxylases (ACCs), rate-limiting enzymes in long chain fatty acid biosynthesis, have greatly advanced the understanding of their biochemistry and molecular biology and promoted the use of ACCs as targets for herbicides in agriculture and for development of drugs for diabetes, obesity and cancers. In mammals, ACCs have both biotin carboxylase (BC) and carboxyltransferase (CT) activity, catalyzing carboxylation of acetyl-CoA to malonyl-CoA. Several classes of small chemicals modulate ACC activity, including cellular metabolites, natural compounds, and chemically synthesized products. This article reviews chemical genetic studies of ACCs and the use of ACCs for targeted therapy of cancers.
Collapse
Affiliation(s)
- Xuyu Zu
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Jing Zhong
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Dixian Luo
- Institute of Translational Medicine & Department of Laboratory Medicine, the First People’s Hospital of Chenzhou, 102 Luojiajing Road, Chenzhou 423000, Hunan, China
| | - Jingjing Tan
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Qinghai Zhang
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Ying Wu
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Jianghua Liu
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Renxian Cao
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
- Authors to whom correspondence should be addressed; E-Mails: (R.C.); (D.C.); Tel.: +86-217-545-9703 (D.C.); Fax: +86-217-545-9718 (D.C.)
| | - Gebo Wen
- Institute of Clinical Medicine, the First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Deliang Cao
- Department of Microbiology, Immunology and Cell Biology, Simmons Cancer Institute, Southern Illinois University School of Medicine, 913 N. Rutledge Street, Springfield, IL 62794, USA
- Authors to whom correspondence should be addressed; E-Mails: (R.C.); (D.C.); Tel.: +86-217-545-9703 (D.C.); Fax: +86-217-545-9718 (D.C.)
| |
Collapse
|
36
|
Cholesterol accumulation in prostate cancer: a classic observation from a modern perspective. Biochim Biophys Acta Rev Cancer 2013; 1835:219-29. [PMID: 23357067 DOI: 10.1016/j.bbcan.2013.01.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/12/2013] [Accepted: 01/15/2013] [Indexed: 12/26/2022]
Abstract
Prostate cancer (PCa) is the most common cancer in men in developed countries. Epidemiological studies have associated high blood-cholesterol levels with an increased risk of PCa, whilst cholesterol-lowering drugs (statins) reduce the risk of advanced PCa. Furthermore, normal prostate epithelial cells have an abnormally high cholesterol content, with cholesterol levels increasing further during progression to PCa. In this review, we explore why and how this occurs. Concurrent to this observation, intense efforts have been expended in cardiovascular research to better understand the regulators of cholesterol homeostasis. Here, we apply this knowledge to elucidate the molecular mechanisms driving the accumulation of cholesterol in PCa. For instance, recent evidence from our group and others shows that major signalling players in prostate growth and differentiation, such as androgens and Akt, modulate the key transcriptional regulators of cholesterol homeostasis to enhance cholesterol levels. This includes adjusting central carbon metabolism to sustain greater lipid synthesis. Perturbations in cholesterol homeostasis appear to be maintained even when PCa approaches the advanced, 'castration-resistant' state. Overall, this provides a link between cholesterol accumulation and PCa cell growth. Given there is currently no cure for castration-resistant PCa, could cholesterol metabolism be a novel target for PCa therapy? Overall, this review presents a picture that cholesterol metabolism is important for PCa development: growth-promoting factors stimulate cholesterol accumulation, which in turn presents a possible target for chemotherapy. Consequently, we recommend future investigations, both to better elucidate the mechanisms driving this accumulation and applying it in novel chemotherapeutic strategies.
Collapse
|
37
|
Yu J, Deng R, Zhu HH, Zhang SS, Zhu C, Montminy M, Davis R, Feng GS. Modulation of fatty acid synthase degradation by concerted action of p38 MAP kinase, E3 ligase COP1, and SH2-tyrosine phosphatase Shp2. J Biol Chem 2012; 288:3823-30. [PMID: 23269672 DOI: 10.1074/jbc.m112.397885] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Src-homology 2 (SH2) domain-containing tyrosine phosphatase Shp2 has been known to regulate various signaling pathways triggered by receptor and cytoplasmic tyrosine kinases. Here we describe a novel function of Shp2 in control of lipid metabolism by mediating degradation of fatty acid synthase (FASN). p38-phosphorylated COP1 accumulates in the cytoplasm and subsequently binds FASN through Shp2 here as an adapter, leading to FASN-Shp2-COP1 complex formation and FASN degradation mediated by ubiquitination pathway. By fasting p38 is activated and stimulates FASN protein degradation in mice. Consistently, the FASN protein levels are dramatically elevated in mouse liver and pancreas in which Shp2/Ptpn11 is selectively deleted. Thus, this study identifies a new activity for Shp2 in lipid metabolism.
Collapse
Affiliation(s)
- Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology & Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fatty acid synthase is a predictive marker for aggressiveness in meningiomas. J Neurooncol 2012; 109:399-404. [PMID: 22744755 DOI: 10.1007/s11060-012-0907-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 05/29/2012] [Indexed: 10/28/2022]
Abstract
Meningiomas are the most frequent intracranial tumors. Although most are benign WHO grade I tumors, grade II and III tumors are aggressive and survival is poor. Treatment options for grade II and III meningiomas are limited, and molecular targets are few. The re-programming of metabolic pathways including glycolysis, lipogenesis, and nucleotide synthesis is a hallmark of the physiological changes in cancer cells. Because fatty acid synthase (FAS), the enzyme responsible for the de-novo synthesis of fatty acids, has emerged as a potential therapeutic target for several cancers, we investigated its involvement in meningiomas. We subjected 92 paraffin-embedded samples from 57 patients with grade I, 18 with grade II and III, and six with radiation-induced tumors to immunohistochemical study of FAS. Whereas its expression was increased in grade II and III meningiomas (62.9 %) compared with grade I tumors (29.8 %) (chi-squared test: p < 0.001), FAS was expressed in grade I tumors with a high MIB-1 index and infiltration into surrounded tissues. All radiation-induced meningiomas expressed FAS and its expression was positively correlated with the MIB-1 index (p < 0.005). Our findings suggest that increased FAS expression reflects the aggressiveness of meningiomas and that it may be a novel therapeutic target for treatment of unresectable or malignant tumors.
Collapse
|
39
|
Fatty acid synthase expression associated with NAC1 is a potential therapeutic target in ovarian clear cell carcinomas. Br J Cancer 2012; 107:300-7. [PMID: 22653145 PMCID: PMC3394978 DOI: 10.1038/bjc.2012.246] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND This study examined the clinical significance of NAC1 and the expression level of its potential downstream target fatty acid synthase (FASN) in ovarian clear cell carcinomas (OCCCs), and evaluated the NAC1/FASN pathway as a potential therapeutic target. METHODS NAC1 and FASN expression and NACC1 gene amplification were assessed in ovarian cancers by immunohistochemistry, fluorescence in situ hybridisation, and clinical data collected by a retrospective chart review. C75, a FASN inhibitor, was used to assess whether this pathway represented a therapeutic target in OCCC. RESULTS High NAC1 expression was most frequent in clear cell tumours (40.0%:24/60). NACC1 gene amplification was identified in none of the 58 OCCCs. The frequency of NACC1 gene amplification was significantly higher in the high-grade serous histology than in the clear cell histology (P<0.01). NAC1 expression was significantly correlated with FASN expression in both OCCC samples and OCCC cell lines. Either high NAC1 expression or high FASN expression significantly correlated with shorter progression-free and overall survival (P=0.002 and 0.0048). NAC1 overexpression stimulated FASN expression, and NAC1 silencing using siRNA decreased FASN expression in OCCC cell lines. Profound growth inhibition was observed in C75-treated carcinoma cells with FASN overexpression when compared with the response in carcinoma cells without FASN expression. CONCLUSION These findings indicate that NAC1/FASN overexpression is critical to the growth and survival of a subset of OCCC. The FASN silencing by the C75-induced phenotypes depends on the expression status of the targeted cell line. Therefore, NAC1/FASN pathway-targeted therapy may benefit selected OCCC patients.
Collapse
|
40
|
Netto GJ, Cheng L. Emerging critical role of molecular testing in diagnostic genitourinary pathology. Arch Pathol Lab Med 2012; 136:372-90. [PMID: 22458900 DOI: 10.5858/arpa.2011-0471-ra] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CONTEXT The unprecedented advances in cancer genetics and genomics are rapidly affecting clinical management and diagnostics in solid tumor oncology. Molecular diagnostics is now an integral part of routine clinical management in patients with lung, colon, and breast cancer. In sharp contrast, molecular biomarkers have been largely excluded from current management algorithms of urologic malignancies. OBJECTIVE To discuss promising candidate biomarkers that may soon make their transition to the realm of clinical management of genitourologic malignancies. The need for new treatment alternatives that can improve upon the modest outcome so far in patients with several types of urologic cancer is evident. Well-validated prognostic molecular biomarkers that can help clinicians identify patients in need of early aggressive management are lacking. Identifying robust predictive biomarkers that will stratify response to emerging targeted therapeutics is another crucially needed development. A compiled review of salient studies addressing the topic could be helpful in focusing future efforts. DATA SOURCES A PubMed (US National Library of Medicine) search for published studies with the following search terms was conducted: molecular , prognostic , targeted therapy , genomics , theranostics and urinary bladder cancer , prostate adenocarcinoma , and renal cell carcinoma . Articles with large cohorts and multivariate analyses were given preference. CONCLUSIONS Our recent understanding of the complex molecular alterations involved in the development and progression of urologic malignancies is yielding novel diagnostic and prognostic molecular tools and opening the doors for experimental targeted therapies for these prevalent, frequently lethal solid tumors.
Collapse
Affiliation(s)
- George J Netto
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
41
|
Moore RG, Lange TS, Robinson K, Kim KK, Uzun A, Horan TC, Kawar N, Yano N, Chu SR, Mao Q, Brard L, DePaepe ME, Padbury JF, Arnold LA, Brodsky A, Shen TL, Singh RK. Efficacy of a non-hypercalcemic vitamin-D2 derived anti-cancer agent (MT19c) and inhibition of fatty acid synthesis in an ovarian cancer xenograft model. PLoS One 2012; 7:e34443. [PMID: 22509304 PMCID: PMC3317945 DOI: 10.1371/journal.pone.0034443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 03/02/2012] [Indexed: 02/07/2023] Open
Abstract
Background Numerous vitamin-D analogs exhibited poor response rates, high systemic toxicities and hypercalcemia in human trials to treat cancer. We identified the first non-hypercalcemic anti-cancer vitamin D analog MT19c by altering the A-ring of ergocalciferol. This study describes the therapeutic efficacy and mechanism of action of MT19c in both in vitro and in vivo models. Methodology/Principal Finding Antitumor efficacy of MT19c was evaluated in ovarian cancer cell (SKOV-3) xenografts in nude mice and a syngenic rat ovarian cancer model. Serum calcium levels of MT19c or calcitriol treated animals were measured. In-silico molecular docking simulation and a cell based VDR reporter assay revealed MT19c–VDR interaction. Genomewide mRNA analysis of MT19c treated tumors identified drug targets which were verified by immunoblotting and microscopy. Quantification of cellular malonyl CoA was carried out by HPLC-MS. A binding study with PPAR-Y receptor was performed. MT19c reduced ovarian cancer growth in xenograft and syngeneic animal models without causing hypercalcemia or acute toxicity. MT19c is a weak vitamin-D receptor (VDR) antagonist that disrupted the interaction between VDR and coactivator SRC2-3. Genome-wide mRNA analysis and western blot and microscopy of MT19c treated xenograft tumors showed inhibition of fatty acid synthase (FASN) activity. MT19c reduced cellular levels of malonyl CoA in SKOV-3 cells and inhibited EGFR/phosphoinositol-3kinase (PI-3K) activity independently of PPAR-gamma protein. Significance Antitumor effects of non-hypercalcemic agent MT19c provide a new approach to the design of vitamin-D based anticancer molecules and a rationale for developing MT19c as a therapeutic agent for malignant ovarian tumors by targeting oncogenic de novo lipogenesis.
Collapse
Affiliation(s)
- Richard G. Moore
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Thilo S. Lange
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Katina Robinson
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Kyu K. Kim
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Alper Uzun
- Center for Computational Molecular Biology, Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Timothy C. Horan
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Nada Kawar
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Naohiro Yano
- Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Sharon R. Chu
- Developmental Pathology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Quanfu Mao
- Developmental Pathology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Laurent Brard
- Gynecology Oncology, Southern Illinois Medical School, Springfield, Illinois, United States of America
| | - Monique E. DePaepe
- Developmental Pathology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - James F. Padbury
- Center for Computational Molecular Biology, Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
| | - Leggy A. Arnold
- Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Alexander Brodsky
- Department of Biology and Medicine, Brown University, Providence, Rhode Island, United States of America
| | - Tun-Li Shen
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Rakesh K. Singh
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Alpert Medical School, Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
42
|
Harris DM, Li L, Chen M, Lagunero FT, Go VLW, Boros LG. Diverse mechanisms of growth inhibition by luteolin, resveratrol, and quercetin in MIA PaCa-2 cells: a comparative glucose tracer study with the fatty acid synthase inhibitor C75. Metabolomics 2012; 8:201-210. [PMID: 22754424 PMCID: PMC3383678 DOI: 10.1007/s11306-011-0300-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The rationale of this dose matching/dose escalating study was to compare a panel of flavonoids-luteolin, resveratrol, and quercetin-against the metabolite flux-controlling properties of a synthetic targeted fatty acid synthase inhibitor drug C75 on multiple macromolecule synthesis pathways in pancreatic tumor cells using [1,2-(13)C(2)]-d-glucose as the single precursor metabolic tracer. MIA PaCa-2 pancreatic adenocarcinoma cells were cultured for 48 h in the presence of 0.1% DMSO (control), or 50 or 100 μM of each test compound, while intracellular glycogen, RNA ribose, palmitate and cholesterol as well as extra cellular (13)CO(2), lactate and glutamate production patterns were measured using gas chromatography/mass spectrometry (GC/MS) and stable isotope-based dynamic metabolic profiling (SiDMAP). The use of 50% [1,2-(13)C(2)]-d-glucose as tracer resulted in an average of 24 excess (13)CO(2) molecules for each 1,000 CO(2) molecule in the culture media, which was decreased by 29 and 33% (P < 0.01) with 100 μM C75 and luteolin treatments, respectively. Extracellular tracer glucose-derived (13)C-labeled lactate fractions (Σm) were between 45.52 and 47.49% in all cultures with a molar ratio of 2.47% M + 1/Σm lactate produced indirectly by direct oxidation of glucose in the pentose cycle in control cultures; treatment with 100 μM C75 and luteolin decreased this figure to 1.80 and 1.67%. The tracer glucose-derived (13)C labeled fraction (Σm) of ribonucleotide ribose was 34.73% in controls, which was decreased to 20.58 and 8.45% with C75, 16.15 and 6.86% with luteolin, 27.66 and 19.25% with resveratrol, and 30.09 and 25.67% with quercetin, respectively. Luteolin effectively decreased nucleotide precursor synthesis pentose cycle flux primarily via the oxidative branch, where we observed a 41.74% flux (M + 1/Σm) in control cells, in comparison with only a 37.19%, 32.74%, or a 26.57%, 25.47% M + 1/Σm flux (P < 0.001) after 50 or 100 μM C75 or luteolin treatment. Intracellular de novo fatty acid palmitate (C16:0) synthesis was severely and equally blocked by C75 and luteolin treatments indicated by the 5.49% (control), 2.29 or 2.47% (C75) and 2.21 or 2.73% (luteolin) tracer glucose-derived (13)C-labeled fractions, respectively. On the other hand there was a significant 192 and 159% (P < 0.001), and a 103 and 117% (P < 0.01) increase in tracer glucose-derived cholesterol after C75 or luteolin treatment. Only resveratrol and quercetin at 100 μM inhibited tracer glucose-derived glycogen labeling (Σm) and turnover by 34.8 and 23.8%, respectively. The flavonoid luteolin possesses equal efficacy to inhibit fatty acid palmitate de novo synthesis as well as nucleotide RNA ribose turnover via the oxidative branch of the pentose cycle in comparison with the targeted fatty acid synthase inhibitor synthetic compound C75. Luteolin is also effective in stringently controlling glucose entry and anaplerosis in the TCA cycle, while it promotes less glucose flux towards cholesterol synthesis than that of C75. In contrast, quercetin and resveratrol inhibit glycogen synthesis and turnover as their underlying mechanism of controlling tumor cell proliferation. Therefore the flavonoid luteolin controls fatty and nucleic acid syntheses as well as energy production with pharmacological strength, which can be explored as a non-toxic natural treatment modality for pancreatic cancer.
Collapse
Affiliation(s)
- Diane M Harris
- Department of Medicine, David Geffen School of Medicine at UCLA, 13-146 Warren Hall, 900 Veteran Ave., Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
43
|
Ros S, Santos CR, Moco S, Baenke F, Kelly G, Howell M, Zamboni N, Schulze A. Functional metabolic screen identifies 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 as an important regulator of prostate cancer cell survival. Cancer Discov 2012; 2:328-43. [PMID: 22576210 DOI: 10.1158/2159-8290.cd-11-0234] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Alterations in metabolic activity contribute to the proliferation and survival of cancer cells. We investigated the effect of siRNA-mediated gene silencing of 222 metabolic enzymes, transporters, and regulators on the survival of 3 metastatic prostate cancer cell lines and a nonmalignant prostate epithelial cell line. This approach revealed significant complexity in the metabolic requirements of prostate cancer cells and identified several genes selectively required for their survival. Among these genes was 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4), an isoform of phosphofructokinase 2 (PFK2). We show that PFKFB4 is required to balance glycolytic activity and antioxidant production to maintain cellular redox balance in prostate cancer cells. Depletion of PFKFB4 inhibited tumor growth in a xenograft model, indicating that it is required under physiologic nutrient levels. PFKFB4 mRNA expression was also found to be greater in metastatic prostate cancer compared with primary tumors. Taken together, these results indicate that PFKFB4 is a potential target for the development of antineoplastic agents. SIGNIFICANCE Cancer cells undergo several changes in their metabolism that promote growth and survival. Using an unbiased functional screen, we found that the glycolytic enzyme PFKFB4 is essential for prostate cancer cell survival by maintaining the balance between the use of glucose for energy generation and the synthesis of antioxidants. Targeting PFKFB4 may therefore present new therapeutic opportunities.
Collapse
Affiliation(s)
- Susana Ros
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
CONTEXT Molecular diagnostic applications are now an integral part of the management algorithms of several solid tumors, such as breast, colon, and lung. In stark contrast, the current clinical management of urologic malignancies is lagging behind. Clinically robust molecular tests that can identify patients who are more likely to respond to a given targeted agent or even those in need of a more aggressive treatment based on well-validated molecular prognosticators are still lacking. Several promising biomarkers for detection, prognosis, and targeted therapeutics are being evaluated. OBJECTIVE To discuss candidate biomarkers that may soon make the transition to clinical assay for patients in urologic oncology. DATA SOURCES Selected original articles published in the PubMed service of the US National Library of Medicine. CONCLUSIONS Recent understanding of the complex molecular alterations involved in the development and progression of urologic malignancies is yielding novel diagnostic and prognostic molecular tools and opening the doors for experimental targeted therapies in these prevalent, frequently lethal solid tumors.
Collapse
Affiliation(s)
- George J Netto
- Department of Pathology, Johns Hopkins Medical Institutions, The Harry and Jeanette Weinberg Building, 401 N Broadway, Baltimore, MD 21231-2410, USA.
| |
Collapse
|
45
|
Krycer JR, Brown AJ. Cross-talk between the androgen receptor and the liver X receptor: implications for cholesterol homeostasis. J Biol Chem 2011; 286:20637-47. [PMID: 21489984 PMCID: PMC3121513 DOI: 10.1074/jbc.m111.227082] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 04/01/2011] [Indexed: 01/16/2023] Open
Abstract
High cholesterol levels are associated with prostate cancer development. Androgens promote cholesterol accumulation by activating the sterol-regulatory element-binding protein isoform 2 (SREBP-2) transcription factor. However, SREBP-2 is in balance with the liver X receptor (LXR; NR1H2/NR1H3), a transcription factor that prevents cholesterol accumulation. Here, we show that LXR activity is down-regulated by the androgen receptor (AR; NR3C4). In turn, this reduces LXR target gene expression. This antagonism on LXR is also exerted by other steroid hormone receptors, including the estrogen, glucocorticoid, and progesterone receptors. This suggests a generalizable mechanism, but the AR does not affect LXR mRNA levels, protein degradation, or DNA binding. We also found that the AR does not require protein synthesis to influence LXR, suggesting a direct antagonism. However, the AR does not directly bind LXR. The AR N-terminal domain (involved in transactivation), but not its DNA-binding domain, is required to suppress LXR activity, suggesting coactivator competition. Overall, this androgen-mediated antagonism of LXR complements SREBP-2 activation, providing a more complete picture as to how androgens increase cellular cholesterol levels in a prostate cancer setting. Given the cross-talk between other steroid hormone receptors and LXR, hormonal regulation of cholesterol via LXR may occur in a variety of cellular contexts.
Collapse
Affiliation(s)
- James Robert Krycer
- From the School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Andrew John Brown
- From the School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
46
|
C11-acetate and F-18 FDG PET for men with prostate cancer bone metastases: relative findings and response to therapy. Clin Nucl Med 2011; 36:192-8. [PMID: 21285676 DOI: 10.1097/rlu.0b013e318208f140] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF THE REPORT This study tested the feasibility of C11-acetate (acetate) positron emission tomography (PET) imaging to assess response to therapy in men with bone metastatic prostate cancer and compared results for disease detection and response evaluation with F-18 fluorodeoxyglucose (FDG) PET. MATERIALS AND METHODS Men with ≥3 prostate cancer bone metastases identified by Tc-99m methylene diphosphonate (MDP) bone scintigraphy and/or computed tomography were enrolled in a prospective study of serial acetate and FDG PET imaging. Patients were imaged before and 6 to 12 weeks after initial androgen deprivation therapy for new metastatic prostate cancer or first-line chemotherapy with docetaxel for castration-resistant prostate cancer. Qualitative assessment and changes in the tumor:normal uptake ratio were used to assess response by both acetate and FDG PET. In addition, the detection of bone metastases pretherapy was compared for acetate and FDG PET. RESULTS A total of 8 patients with documented bone metastases were imaged, of which 6 were imaged both pre- and post-therapy. Acetate PET detected bone metastases in all 8 patients, whereas FDG PET detected lesions in 6 of the 7 imaged patients. Acetate PET generally detected more metastases with a higher tumor:normal uptake ratio. Qualitative and quantitative assessments of post-treatment response correlated with composite clinical designations of response, stable disease, or progression in 6 of 6 and 5 of 6 by acetate and 4 of 5 and 3 of 5 by FDG PET, respectively. CONCLUSIONS In this pilot study, results indicate that acetate PET holds promise for response assessment of prostate cancer bone metastases and is complementary to FDG PET in bone metastasis detection.
Collapse
|
47
|
Uddin S, Jehan Z, Ahmed M, Alyan A, Al-Dayel F, Hussain A, Bavi P, Al-Kuraya KS. Overexpression of fatty acid synthase in Middle Eastern epithelial ovarian carcinoma activates AKT and Its inhibition potentiates cisplatin-induced apoptosis. Mol Med 2011; 17:635-45. [PMID: 21442130 DOI: 10.2119/molmed.2011.00046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 03/16/2011] [Indexed: 12/24/2022] Open
Abstract
Fatty acid synthase (FASN), the enzyme responsible for de novo synthesis of fatty acids, has been shown to be deregulated in several cancers, including epithelial ovarian carcinoma (EOC). In this study, we investigated the function of the FASN signaling pathway in a large series of Middle Eastern EOC patient samples, a panel of cell lines and nude mouse model. Using immunohistochemistry, we detected overexpression of FASN in 75.5% (114/151) of the tumor samples. Overexpression of FASN was associated significantly with tumor proliferative marker Ki-67 (P = 0.0009), activated AKT (P = 0.0117) and XIAP (P = 0.0046). Treatment of EOC cell lines with C-75, a selective inhibitor of FASN, caused inhibition of EOC cell viability via induction of apoptosis. Inhibition of FASN by C-75 led apoptosis via the mitochondrial pathway. FASN inhibition caused downregulation of activated AKT and its downstream targets. In addition, inhibition by FASN siRNA caused downregulation of FASN and activation of caspases, suggesting the role of FASN in C-75 mediated apoptosis. Furthermore, treatment of EOC cells with subtoxic doses of C-75 augmented the effect of cisplatin-mediated induction of apoptosis. Finally, treatment of EOC cell line xenografts with a combination of C-75 and cisplatin resulted in growth inhibition of tumors in nude mice through downregulation of FASN and activation of caspases. Altogether, our results show overexpression of FASN in Middle Eastern EOC, suggesting that FASN may be a potential therapeutic target in a subset of EOC, alone or in combination with other conventional chemotherapeutic agents.
Collapse
Affiliation(s)
- Shahab Uddin
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Silva SD, Cunha IW, Younes RN, Soares FA, Kowalski LP, Graner E. ErbB receptors and fatty acid synthase expression in aggressive head and neck squamous cell carcinomas. Oral Dis 2011; 16:774-80. [PMID: 20604875 DOI: 10.1111/j.1601-0825.2010.01687.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
SUMMARY Overexpression of ErbB receptors is frequent in head and neck squamous cell carcinomas (HNSCC) and seems to be correlated with tumor progression and metastasis. Fatty acid synthase (FASN), the key lipogenic enzyme responsible for the endogenous synthesis of fatty acids, is regulated by ErbB2 and overexpressed in several human malignancies. METHODS This study was performed to examine the immunohistochemical expression patterns of ErbB1, ErbB2, ErbB3, ErbB4, and FASN in a tissue microarray, containing 33 representative areas from aggressive primary HNSCC (whose patients had distant metastasis), and 21 matched lung metastasis. RESULTS Strong correlation among the expression of ErbB family receptors was found (ErbB1-ErbB2 P = 0.008, ErbB1-ErbB4 P = 0.018, EbB2-ErbB3 P = 0.001, ErbB2-ErbB4 P = 0.006, ErbB3-ErbB4 P=0.012) in the HNSCC. FASN expression was significantly associated with ErbB2 (P = 0.024). Lymphatic permeation was correlated with ErbB3 (P = 0.033) and histological grade with ErbB4 staining (P = 0.050). ErbB1 and ErbB2 were found mainly in patients with smoking habit (P = 0.011 and P = 0.027), and ErbB2 was associated with alcohol consumption and clinical stage (P = 0.014 and P = 0.031). Finally, FASN was overexpressed in lung metastasis, in comparison with matched HNSCC samples (P = 0.006). CONCLUSIONS The results showed that high FASN immunohistochemical expression is a feature of HNSCC lung metastasis, and ErbB1-ErbB2, ErbB1-ErbB4, ErbB2-ErbB3, ErbB2-ErbB4, and ErbB3-ErbB4 expression levels are correlated in the respective primary tumors, being ErbB2 the preferred coexpression partner of all the other ErbB receptors.
Collapse
Affiliation(s)
- S D Silva
- Department of Head and Neck Surgery and Otorhinolaryngology Anatomic Pathology Thoracic Surgery, A.C. Camargo Hospital, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
49
|
Nguyen PL, Ma J, Chavarro JE, Freedman ML, Lis R, Fedele G, Fiore C, Qiu W, Fiorentino M, Finn S, Penney KL, Eisenstein A, Schumacher FR, Mucci LA, Stampfer MJ, Giovannucci E, Loda M. Fatty acid synthase polymorphisms, tumor expression, body mass index, prostate cancer risk, and survival. J Clin Oncol 2010; 28:3958-64. [PMID: 20679621 PMCID: PMC2940394 DOI: 10.1200/jco.2009.27.0793] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 06/21/2010] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Fatty acid synthase (FASN) regulates de novo lipogenesis, body weight, and tumor growth. We examined whether common germline single nucleotide polymorphisms (SNPs) in the FASN gene affect prostate cancer (PCa) risk or PCa-specific mortality and whether these effects vary by body mass index (BMI). METHODS In a prospective nested case-control study of 1,331 white patients with PCa and 1,267 age-matched controls, we examined associations of five common SNPs within FASN (and 5 kb upstream/downstream, R(2) > 0.8) with PCa incidence and, among patients, PCa-specific death and tested for an interaction with BMI. Survival analyses were repeated for tumor FASN expression (n = 909). RESULTS Four of the five SNPs were associated with lethal PCa. SNP rs1127678 was significantly related to higher BMI and interacted with BMI for both PCa risk (P(interaction) = .004) and PCa mortality (P(interaction) = .056). Among overweight men (BMI > or = 25 kg/m(2)), but not leaner men, the homozygous variant allele carried a relative risk of advanced PCa of 2.49 (95% CI, 1.00 to 6.23) compared with lean men with the wild type. Overweight patients carrying the variant allele had a 2.04 (95% CI, 1.31 to 3.17) times higher risk of PCa mortality. Similarly, overweight patients with elevated tumor FASN expression had a 2.73 (95% CI, 1.05 to 7.08) times higher risk of lethal PCa (P(interaction) = .02). CONCLUSION FASN germline polymorphisms were significantly associated with risk of lethal PCa. Significant interactions of BMI with FASN polymorphisms and FASN tumor expression suggest FASN as a potential link between obesity and poor PCa outcome and raise the possibility that FASN inhibition could reduce PCa-specific mortality, particularly in overweight men.
Collapse
Affiliation(s)
- Paul L. Nguyen
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Jing Ma
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Jorge E. Chavarro
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Matthew L. Freedman
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Rosina Lis
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Giuseppe Fedele
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Christopher Fiore
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Weiliang Qiu
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Michelangelo Fiorentino
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Stephen Finn
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Kathryn L. Penney
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Anna Eisenstein
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Fredrick R. Schumacher
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Lorelei A. Mucci
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Meir J. Stampfer
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Edward Giovannucci
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Massimo Loda
- From the Dana-Farber Cancer Institute; Brigham & Women's Hospital; Children's Hospital Boston; Harvard School of Public Health, Harvard Medical School, Boston; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; and University of Southern California Keck School of Medicine, Los Angeles, CA
| |
Collapse
|
50
|
Murata S, Yanagisawa K, Fukunaga K, Oda T, Kobayashi A, Sasaki R, Ohkohchi N. Fatty acid synthase inhibitor cerulenin suppresses liver metastasis of colon cancer in mice. Cancer Sci 2010; 101:1861-5. [PMID: 20491775 PMCID: PMC11159773 DOI: 10.1111/j.1349-7006.2010.01596.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Fatty acid synthase (FAS) is highly expressed in many kinds of human cancers, including colorectal cancer (CRC), and we have investigated the potential use of FAS inhibitors for chemoprevention of liver metastasis of CRC in mice. Expression of FAS was evaluated in murine CRC cell lines Colon 26 and CMT 93. Cerulenin, a natural inhibitor of FAS, induced apoptosis in these cell lines. The ability of cerulenin to prevent development of liver metastatic lesions in Colon 26 was evaluated. The numbers and sizes of liver metastatic CRC tumors were significantly reduced by treating mice with cerulenin. Cerulenin treatment was associated with reduced levels of phosphorylated Akt in Colon 26 cells, suggesting that inhibition of this signal transduction pathway might be involved in the chemopreventive activity of this compound. Based on studies in mouse models, inhibiting FAS would be an effective strategy to prevent and retard growth of liver metastatic tumors of CRC that have high expression of this enzyme.
Collapse
Affiliation(s)
- Soichiro Murata
- Department of Surgery, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | | | | | |
Collapse
|