1
|
Meddeb M, Koleini N, Jun S, Keykhaei M, Farshidfar F, Zhao L, Kwon S, Lin B, Keceli G, Paolocci N, Hahn V, Sharma K, Pearce EL, Kass DA. ATP Citrate Lyase Supports Cardiac Function and NAD+/NADH Balance And Is Depressed in Human Heart Failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598152. [PMID: 38915649 PMCID: PMC11195057 DOI: 10.1101/2024.06.09.598152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND ATP-citrate lyase (ACLY) converts citrate into acetyl-CoA and oxaloacetate in the cytosol. It plays a prominent role in lipogenesis and fat accumulation coupled to excess glucose, and its inhibition is approved for treating hyperlipidemia. In RNAseq analysis of human failing myocardium, we found ACLY gene expression is reduced; however the impact this might have on cardiac function and/or metabolism has not been previously studied. As new ACLY inhibitors are in development for cancer and other disorders, such understanding has added importance. METHODS Cardiomyocytes, ex-vivo beating hearts, and in vivo hearts with ACLY inhibited by selective pharmacologic (BMS303141, ACLYi) or genetic suppression, were studied. Regulation of ACLY gene/protein expression, and effects of ACLYi on function, cytotoxicity, tricarboxylic acid (TCA)-cycle metabolism, and redox and NAD+/NADH balance were assessed. Mice with cardiac ACLY knockdown induced by AAV9-acly-shRNA or cardiomyocyte tamoxifen-inducible Acly knockdown were studied. RESULTS Acly gene expression was reduced more in obese patients with heart failure and preserved EF (HFpEF) than HF with reduced EF. In vivo pressure-overload and in vitro hormonal stress increased ACLY protein expression, whereas it declined upon fatty-acid exposure. Acute ACLYi (1-hr) dose-dependently induced cytotoxicity in adult and neonatal cardiomyocytes, and caused substantial reduction of systolic and diastolic function in myocytes and ex-vivo beating hearts. In the latter, ATP/ADP ratio also fell and lactate increased. U13C-glucose tracing revealed an ACLYdependent TCA-bypass circuit in myocytes, where citrate generated in mitochondria is transported to the cytosol, metabolized by ACLY and then converted to malate to re-enter mitochondria,bypassing several NADH-generating steps. ACLYi lowered NAD+/NADH ratio and restoring this balance ameliorated cardiomyocyte toxicity. Oxidative stress was undetected with ACLYi. Adult hearts following 8-weeks of reduced cardiac and/or cardiomyocyte ACLY downregulation exhibited ventricular dilation and reduced function that was prevented by NAD augmentation. Cardiac dysfunction from ACLY knockdown was worse in hearts subjected to sustained pressureoverload, supporting a role in stress responses. CONCLUSIONS ACLY supports normal cardiac function through maintenance of the NAD+/NADH balance and is upregulated by hemodynamic and hormonal stress, but depressed by lipid excess. ACLY levels are most reduced in human HFpEF with obesity potentially worsening cardio-metabolic reserve.
Collapse
|
2
|
Chung HY, Lee GS, Nam SH, Lee JH, Han JP, Song S, Kim GD, Jung C, Hyeon DY, Hwang D, Choi BO, Yeom SC. Morc2a variants cause hydroxyl radical-mediated neuropathy and are rescued by restoring GHKL ATPase. Brain 2024; 147:2114-2127. [PMID: 38227798 DOI: 10.1093/brain/awae017] [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: 07/31/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Mutations in the Microrchidia CW-type zinc finger 2 (MORC2) GHKL ATPase module cause a broad range of neuropathies, such as Charcot-Marie-Tooth disease type 2Z; however, the aetiology and therapeutic strategy are not fully understood. Previously, we reported that the Morc2a p.S87L mouse model exhibited neuropathy and muscular dysfunction through DNA damage accumulation. In the present study, we analysed the gene expression of Morc2a p.S87L mice and designated the primary causing factor. We investigated the pathological pathway using Morc2a p.S87L mouse embryonic fibroblasts and human fibroblasts harbouring MORC2 p.R252W. We subsequently assessed the therapeutic effect of gene therapy administered to Morc2a p.S87L mice. This study revealed that Morc2a p.S87L causes a protein synthesis defect, resulting in the loss of function of Morc2a and high cellular apoptosis induced by high hydroxyl radical levels. We considered the Morc2a GHKL ATPase domain as a therapeutic target because it simultaneously complements hydroxyl radical scavenging and ATPase activity. We used the adeno-associated virus (AAV)-PHP.eB serotype, which has a high CNS transduction efficiency, to express Morc2a or Morc2a GHKL ATPase domain protein in vivo. Notably, AAV gene therapy ameliorated neuropathy and muscular dysfunction with a single treatment. Loss-of-function characteristics due to protein synthesis defects in Morc2a p.S87L were also noted in human MORC2 p.S87L or p.R252W variants, indicating the correlation between mouse and human pathogenesis. In summary, CMT2Z is known as an incurable genetic disorder, but the present study demonstrated its mechanisms and treatments based on established animal models. This study demonstrates that the Morc2a p.S87L variant causes hydroxyl radical-mediated neuropathy, which can be rescued through AAV-based gene therapy.
Collapse
Affiliation(s)
- Hye Yoon Chung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Geon Seong Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Soo Hyun Nam
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
| | - Jeong Hyeon Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Jeong Pil Han
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Sumin Song
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Gap-Don Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Choonkyun Jung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Bioinformatics Institute, Bio-MAX, Seoul National University, Seoul 08826, Republic of Korea
| | - Byung-Ok Choi
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
- Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul 06351, Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
- Department of Agricultural Biotechnology, WCU Biomodulation Major, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
3
|
Liang JJ, Zhou XF, Long H, Li CY, Wei J, Yu XQ, Guo ZY, Zhou YQ, Deng ZS. Recent advance of ATP citrate lyase inhibitors for the treatment of cancer and related diseases. Bioorg Chem 2024; 142:106933. [PMID: 37890210 DOI: 10.1016/j.bioorg.2023.106933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023]
Abstract
ATP citrate lyase (ACLY), a strategic metabolic enzyme that catalyzes the glycolytic to lipidic metabolism, has gained increasing attention as an attractive therapeutic target for hyperlipidemia, cancers and other human diseases. Despite of continual research efforts, targeting ACLY has been very challenging. In this field, most reported ACLY inhibitors are "substrate-like" analogues, which occupied with the same active pockets. Besides, some ACLY inhibitors have been disclosed through biochemical screening or high throughput virtual screening. In this review, we briefly summarized the cancer-related functions and the recent advance of ACLY inhibitors with a particular focus on the SAR studies and their modes of action. We hope to provide a timely and updated overview of ACLY and the discovery of new ACLY inhibitors.
Collapse
Affiliation(s)
- Jian-Jia Liang
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xiang-Feng Zhou
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hui Long
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Chun-Yun Li
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jing Wei
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xiao-Qin Yu
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Zhi-Yong Guo
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yi-Qing Zhou
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhang-Shuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, Key Laboratory of Functional Yeast, China National Light Industry, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
4
|
Kim DH, Song NY, Yim H. Targeting dysregulated lipid metabolism in the tumor microenvironment. Arch Pharm Res 2023; 46:855-881. [PMID: 38060103 PMCID: PMC10725365 DOI: 10.1007/s12272-023-01473-y] [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: 09/27/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
The reprogramming of lipid metabolism and its association with oncogenic signaling pathways within the tumor microenvironment (TME) have emerged as significant hallmarks of cancer. Lipid metabolism is defined as a complex set of molecular processes including lipid uptake, synthesis, transport, and degradation. The dysregulation of lipid metabolism is affected by enzymes and signaling molecules directly or indirectly involved in the lipid metabolic process. Regulation of lipid metabolizing enzymes has been shown to modulate cancer development and to avoid resistance to anticancer drugs in tumors and the TME. Because of this, understanding the metabolic reprogramming associated with oncogenic progression is important to develop strategies for cancer treatment. Recent advances provide insight into fundamental mechanisms and the connections between altered lipid metabolism and tumorigenesis. In this review, we explore alterations to lipid metabolism and the pivotal factors driving lipid metabolic reprogramming, which exacerbate cancer progression. We also shed light on the latest insights and current therapeutic approaches based on small molecular inhibitors and phytochemicals targeting lipid metabolism for cancer treatment. Further investigations are worthwhile to fully understand the underlying mechanisms and the correlation between altered lipid metabolism and carcinogenesis.
Collapse
Affiliation(s)
- Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, 16227, Korea
| | - Na-Young Song
- Department of Applied Life Science, The Graduate School, BK21 Four Project, Yonsei University, Seoul, 03722, Korea
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Korea.
| |
Collapse
|
5
|
Tyutereva EV, Dalinova AA, Demchenko KN, Dmitrieva VA, Dubovik VR, Lukinskiy YV, Mitina GV, Voitsekhovskaja OV, Berestetskiy A. Effects of Phytotoxic Nonenolides, Stagonolide A and Herbarumin I, on Physiological and Biochemical Processes in Leaves and Roots of Sensitive Plants. Toxins (Basel) 2023; 15:toxins15040234. [PMID: 37104172 PMCID: PMC10145764 DOI: 10.3390/toxins15040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/18/2023] [Indexed: 04/28/2023] Open
Abstract
Phytotoxic macrolides attract attention as prototypes of new herbicides. However, their mechanisms of action (MOA) on plants have not yet been elucidated. This study addresses the effects of two ten-membered lactones, stagonolide A (STA) and herbarumin I (HBI) produced by the fungus Stagonospora cirsii, on Cirsium arvense, Arabidopsis thaliana and Allium cepa. Bioassay of STA and HBI on punctured leaf discs of C. arvense and A. thaliana was conducted at a concentration of 2 mg/mL to evaluate phenotypic responses, the content of pigments, electrolyte leakage from leaf discs, the level of reactive oxygen species, Hill reaction rate, and the relative rise in chlorophyll a fluorescence. The toxin treatments resulted in necrotic and bleached leaf lesions in the dark and in the light, respectively. In the light, HBI treatment caused the drop of carotenoids content in leaves on both plants. The electrolyte leakage caused by HBI was light-dependent, in contrast with that caused by STA. Both compounds induced light-independent peroxide generation in leaf cells but did not affect photosynthesis 6 h after treatment. STA (10 µg/mL) caused strong disorders in root cells of A. thaliana leading to the complete dissipation of the mitochondrial membrane potential one hour post treatment, as well as DNA fragmentation and disappearance of acidic vesicles in the division zone after 8 h; the effects of HBI (50 µg/mL) were much milder. Furthermore, STA was found to inhibit mitosis but did not affect the cytoskeleton in cells of root tips of A. cepa and C. arvense, respectively. Finally, STA was supposed to inhibit the intracellular vesicular traffic from the endoplasmic reticulum to the Golgi apparatus, thus interfering with mitosis. HBI is likely to have another main MOA, probably inhibiting the biosynthesis of carotenoids.
Collapse
Affiliation(s)
- Elena V Tyutereva
- Laboratory of Molecular and Ecological Physiology, Komarov Botanical Institute, Russian Academy of Sciences, 197022 Saint-Petersburg, Russia
| | - Anna A Dalinova
- Laboratory of Phytotoxicology and Biotechnology, All-Russian Institute of Plant Protection, Pushkin, 196608 Saint-Petersburg, Russia
| | - Kirill N Demchenko
- Laboratory of Cellular and Molecular Mechanisms of Plant Development, Komarov Botanical Institute, Russian Academy of Sciences, 197022 Saint-Petersburg, Russia
| | - Valeriya A Dmitrieva
- Laboratory of Molecular and Ecological Physiology, Komarov Botanical Institute, Russian Academy of Sciences, 197022 Saint-Petersburg, Russia
| | - Vsevolod R Dubovik
- Laboratory of Phytotoxicology and Biotechnology, All-Russian Institute of Plant Protection, Pushkin, 196608 Saint-Petersburg, Russia
| | - Yuriy V Lukinskiy
- Laboratory of Molecular and Ecological Physiology, Komarov Botanical Institute, Russian Academy of Sciences, 197022 Saint-Petersburg, Russia
- Laboratory of Phytotoxicology and Biotechnology, All-Russian Institute of Plant Protection, Pushkin, 196608 Saint-Petersburg, Russia
| | - Galina V Mitina
- Laboratory of Microbiological Plant Protection, All-Russian Institute of Plant Protection, Pushkin, 196608 Saint-Petersburg, Russia
| | - Olga V Voitsekhovskaja
- Laboratory of Molecular and Ecological Physiology, Komarov Botanical Institute, Russian Academy of Sciences, 197022 Saint-Petersburg, Russia
| | - Alexander Berestetskiy
- Laboratory of Phytotoxicology and Biotechnology, All-Russian Institute of Plant Protection, Pushkin, 196608 Saint-Petersburg, Russia
| |
Collapse
|
6
|
Velez BC, Petrella CP, DiSalvo KH, Cheng K, Kravtsov R, Krasniqi D, Krucher NA. Combined inhibition of ACLY and CDK4/6 reduces cancer cell growth and invasion. Oncol Rep 2023; 49:32. [PMID: 36562384 PMCID: PMC9827262 DOI: 10.3892/or.2022.8469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The use of small molecule kinase inhibitors, which target specific enzymes that are overactive in cancer cells, has revolutionized cancer patient treatment. To treat some types of breast cancer, CDK4/6 inhibitors, such as palbociclib, have been developed that target the phosphorylation of the retinoblastoma tumor suppressor gene. Acquired resistance to CDK4/6 inhibitors may be due to activation of the AKT pro‑survival signaling pathway that stimulates several processes, such as growth, metastasis and changes in metabolism that support rapid cell proliferation. The aim of the present study was to investigate whether targeting ATP citrate lyase (ACLY), a downstream target of AKT, may combine with CDK4/6 inhibition to inhibit tumorigenesis. The present study determined that ACLY is activated in breast and pancreatic cancer cells in response to palbociclib treatment and AKT mediates this effect. Inhibition of ACLY using bempedoic acid used in combination with palbociclib reduced cell viability in a panel of breast and pancreatic cancer cell lines. This effect was also observed using breast cancer cells grown in 3D cell culture. Mechanistically, palbociclib inhibited cell proliferation, whereas bempedoic acid stimulated apoptosis. Finally, using Transwell invasion assays and immunoblotting, the present study demonstrated that ACLY inhibition blocked cell invasion, when used alone or in combination with palbociclib. These data may yield useful information that could guide the development of future therapies aimed at the reduction of acquired resistance observed clinically.
Collapse
Affiliation(s)
| | | | | | - Keyi Cheng
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Rebecca Kravtsov
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Dorina Krasniqi
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| | - Nancy Ann Krucher
- Department of Biology, Pace University, Pleasantville, NY 10570, USA
| |
Collapse
|
7
|
Jovankić JV, Nikodijević DD, Milutinović MG, Nikezić AG, Kojić VV, Cvetković AM, Cvetković DM. Potential of Orlistat to induce apoptotic and antiangiogenic effects as well as inhibition of fatty acid synthesis in breast cancer cells. Eur J Pharmacol 2023; 939:175456. [PMID: 36528070 DOI: 10.1016/j.ejphar.2022.175456] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Breast cancer as most often women's cancer is the second cause of mortality worldwide. Research interest increased in testing non-standard drugs to suppress breast cancer progression and become significant supplements in anticancer therapy. The anti-obesity drug Orlistat showed significant ability for modulation of cancer cell metabolism via antiproliferative, proapoptotic, antiangiogenic, antimetastatic, and hypolipidemic effects. The anticancer potential of Orlistat was evaluated by cytotoxicity (MTT assay), type of cell death (AO/EB double staining), determination of redox status parameters (superoxide, hydrogen peroxide, lipid peroxidation, reduced glutathione), and total lipid levels with colorimetric methods, as well on angiogenesis-related (VEGF, MMP-9, CXCR4/CXCL12) and fatty acid synthesis-related (ACLY, ACC, FASN) parameters on gene and protein levels (immunocytochemistry and qPCR). Based on obtained results Orlistat induces significant cytotoxic, proapoptotic, and anti-angiogenic effects in MDA-MB-231, MDA-MB-468 and MCF-7 breast cancer cells, without significant cytotoxic effects on normal MRC-5 cells. It decreased total lipid levels and changed redox status parameters and cancer cell metabolism via suppression of genes and proteins involved and fatty acid synthesis. Based on showed, Orlistat may be an important supplement in antiangiogenic therapy against breast cancer with no side effects on normal cells, making it a good candidate for future clinical trials.
Collapse
Affiliation(s)
- Jovana V Jovankić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Danijela D Nikodijević
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Milena G Milutinović
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia.
| | - Aleksandra G Nikezić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Vesna V Kojić
- University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr Goldmana 4, Sremska Kamenica, 21204, Serbia
| | - Aleksandar M Cvetković
- University of Kragujevac, Faculty of Medical Sciences, Department of Surgery, Svetozara Markovića 69, 34000, Kragujevac, Serbia
| | - Danijela M Cvetković
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| |
Collapse
|
8
|
Hydroxycitric Acid Inhibits Chronic Myelogenous Leukemia Growth through Activation of AMPK and mTOR Pathway. Nutrients 2022; 14:nu14132669. [PMID: 35807850 PMCID: PMC9268148 DOI: 10.3390/nu14132669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic regulation of cancer cell growth via AMP-activated protein kinase (AMPK) activation is a widely studied strategy for cancer treatment, including leukemias. Recent notions that naturally occurring compounds might have AMPK activity led to the search for nutraceuticals with potential AMPK-stimulating activity. We found that hydroxycitric acid (HCA), a natural, safe bioactive from the plant Garcinia gummi-gutta (cambogia), has potent AMPK activity in chronic myelogenous leukemia (CML) cell line K562. HCA is a known competitive inhibitor of ATP citrate lyase (ACLY) and is widely used as a weight loss inducer. We found that HCA was able to inhibit the growth of K562 cells in in vitro and in vivo xenograft models. At the mechanistic level, we identified a direct interaction between AMPK and ACLY that seems to be sensitive to HCA treatment. Additionally, HCA treatment resulted in the co-activation of AMPK and the mammalian target of rapamycin (mTOR) pathways. Moreover, we found an enhanced unfolded protein response as observed by activation of the eIF2α/ATF4 pathway that could explain the induction of cell cycle arrest at the G2/M phase and DNA fragmentation upon HCA treatment in K562 cells. Overall, these findings suggest HCA as a nutraceutical approach for the treatment of CMLs.
Collapse
|
9
|
Over-Reduced State of Mitochondria as a Trigger of "β-Oxidation Shuttle" in Cancer Cells. Cancers (Basel) 2022; 14:cancers14040871. [PMID: 35205619 PMCID: PMC8870273 DOI: 10.3390/cancers14040871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
A considerable amount of data have accumulated in the last decade on the pronounced mitochondrial fatty acid oxidation (mFAO) in many types of cancer cells. As a result, mFAO was found to coexist with abnormally activated fatty acid synthesis (FAS) and the mevalonate pathway. Recent studies have demonstrated that overactivated mitochondrial β-oxidation may aggravate the impaired mitochondrial redox state and vice versa. Furthermore, the impaired redox state of cancerous mitochondria can ensure the continuous operation of β-oxidation by disconnecting it from the Krebs cycle and connecting it to the citrate-malate shuttle. This could create a new metabolic state/pathway in cancer cells, which we have called the "β-oxidation-citrate-malate shuttle", or "β-oxidation shuttle" for short, which forces them to proliferate. The calculation of the phosphate/oxygen ratio indicates that it is inefficient as an energy source and must consume significantly more oxygen per mole of ATP produced when combined with acetyl-CoA consuming pathways, such as the FAS and mevalonate pathways. The "β-oxidation shuttle" is an unconventional mFAO, a separate metabolic pathway that has not yet been explored as a source of energy, as well as a source of cataplerosis, leading to biomass accumulation, accelerated oxygen consumption, and, ultimately, a source of proliferation. The role of the "β-oxidation shuttle" and its contribution to redox-altered cancer metabolism provides a new direction for the development of future anticancer strategies. This may represent the metabolic "secret" of cancer underlying hypoxia and genomic instability.
Collapse
|
10
|
A “Weird” Mitochondrial Fatty Acid Oxidation as a Metabolic “Secret” of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2339584. [PMID: 35178152 PMCID: PMC8847026 DOI: 10.1155/2022/2339584] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/29/2021] [Indexed: 12/15/2022]
Abstract
Cancer metabolism is an extensively studied field since the discovery of the Warburg effect about 100 years ago and continues to be increasingly intriguing and enigmatic so far. It has become clear that glycolysis is not the only abnormally activated metabolic pathway in the cancer cells, but the same is true for the fatty acid synthesis (FAS) and mevalonate pathway. In the last decade, a lot of data have been accumulated on the pronounced mitochondrial fatty acid oxidation (mFAO) in many types of cancer cells. In this article, we discuss how mFAO can escape normal regulation under certain conditions and be overactivated. Such abnormal activation of mitochondrial β-oxidation can also be combined with mutations in certain enzymes of the Krebs cycle that are common in cancer. If overactivated β-oxidation is combined with other common cancer conditions, such as dysfunctions in the electron transport complexes, and/or hypoxia, this may alter the redox state of the mitochondrial matrix. We propose the idea that the altered mitochondrial redox state and/or inhibited Krebs cycle at certain segments may link mitochondrial β-oxidation to the citrate-malate shuttle instead to the Krebs cycle. We call this abnormal metabolic condition “β-oxidation shuttle”. It is unconventional mFAO, a separate metabolic pathway, unexplored so far as a source of energy, as well as a source of cataplerosis, leading to biomass accumulation, accelerated oxygen consumption, and ultimately a source of proliferation. It is inefficient as an energy source and must consume significantly more oxygen per mole of ATP produced when combined with acetyl-CoA consuming pathways, such as the FAS and mevalonate pathway.
Collapse
|
11
|
Dong W, Liu X, Yang C, Wang D, Xue Y, Ruan X, Zhang M, Song J, Cai H, Zheng J, Liu Y. Glioma glycolipid metabolism: MSI2-SNORD12B-FIP1L1-ZBTB4 feedback loop as a potential treatment target. Clin Transl Med 2021; 11:e411. [PMID: 34047477 PMCID: PMC8114150 DOI: 10.1002/ctm2.411] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
Abnormal energy metabolism, including enhanced aerobic glycolysis and lipid synthesis, is a well-established feature of glioblastoma (GBM) cells. Thus, targeting the cellular glycolipid metabolism can be a feasible therapeutic strategy for GBM. This study aimed to evaluate the roles of MSI2, SNORD12B, and ZBTB4 in regulating the glycolipid metabolism and proliferation of GBM cells. MSI2 and SNORD12B expression was significantly upregulated and ZBTB4 expression was significantly low in GBM tissues and cells. Knockdown of MSI2 or SNORD12B or overexpression of ZBTB4 inhibited GBM cell glycolipid metabolism and proliferation. MSI2 may improve SNORD12B expression by increasing its stability. Importantly, SNORD12B increased utilization of the ZBTB4 mRNA transcript distal polyadenylation signal in alternative polyadenylation processing by competitively combining with FIP1L1, which decreased ZBTB4 expression because of the increased proportion of the 3' untranslated region long transcript. ZBTB4 transcriptionally suppressed the expression of HK2 and ACLY by binding directly to the promoter regions. Additionally, ZBTB4 bound the MSI promoter region to transcriptionally suppress MSI2 expression, thereby forming an MSI2/SNORD12B/FIP1L1/ZBTB4 feedback loop to regulate the glycolipid metabolism and proliferation of GBM cells. In conclusion, MSI2 increased the stability of SNORD12B, which regulated ZBTB4 alternative polyadenylation processing by competitively binding to FIP1L1. Thus, the MSI2/SNORD12B/FIP1L1/ZBTB4 positive feedback loop plays a crucial role in regulating the glycolipid metabolism of GBM cells and provides a potential drug target for glioma treatment.
Collapse
Affiliation(s)
- Weiwei Dong
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Xiaobai Liu
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Chunqing Yang
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Di Wang
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Yixue Xue
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangChina
- Key Laboratory of Cell Biology, Ministry of Public Health of ChinaChina Medical UniversityShenyangChina
- Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaChina Medical UniversityShenyangChina
| | - Xuelei Ruan
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangChina
- Key Laboratory of Cell Biology, Ministry of Public Health of ChinaChina Medical UniversityShenyangChina
- Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaChina Medical UniversityShenyangChina
| | - Mengyang Zhang
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangChina
- Key Laboratory of Cell Biology, Ministry of Public Health of ChinaChina Medical UniversityShenyangChina
- Key Laboratory of Medical Cell Biology, Ministry of Education of ChinaChina Medical UniversityShenyangChina
| | - Jian Song
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Heng Cai
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Jian Zheng
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| | - Yunhui Liu
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Neuro‐oncology in Liaoning ProvinceShenyangChina
- Liaoning Province Medical Surgery and Rehabilitation Robot Technology Engineering Research CenterShenyangChina
| |
Collapse
|
12
|
Gözen D, Kahraman DC, Narci K, Shehwana H, Konu Ö, Çetin-Atalay R. Transcriptome profiles associated with selenium-deficiency-dependent oxidative stress identify potential diagnostic and therapeutic targets in liver cancer cells. ACTA ACUST UNITED AC 2021; 45:149-161. [PMID: 33907497 PMCID: PMC8068766 DOI: 10.3906/biy-2009-56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/01/2021] [Indexed: 12/09/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancer types with high mortality rates and displays increased resistance to various stress conditions such as oxidative stress. Conventional therapies have low efficacies due to resistance and off-target effects in HCC. Here we aimed to analyze oxidative stress-related gene expression profiles of HCC cells and identify genes that could be crucial for novel diagnostic and therapeutic strategies. To identify important genes that cause resistance to reactive oxygen species (ROS), a model of oxidative stress upon selenium (Se) deficiency was utilized. The results of transcriptome-wide gene expression data were analyzed in which the differentially expressed genes (DEGs) were identified between HCC cell lines that are either resistant or sensitive to Se-deficiency-dependent oxidative stress. These DEGs were further investigated for their importance in oxidative stress resistance by network analysis methods, and 27 genes were defined to have key roles; 16 of which were previously shown to have impact on liver cancer patient survival. These genes might have Se-deficiency-dependent roles in hepatocarcinogenesis and could be further exploited for their potentials as novel targets for diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Damla Gözen
- Cancer Systems Biology Laboratory, Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara Turkey
| | - Deniz Cansen Kahraman
- Cancer Systems Biology Laboratory, Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara Turkey
| | - Kübra Narci
- Cancer Systems Biology Laboratory, Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara Turkey
| | - Huma Shehwana
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi Pakistan
| | - Özlen Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara Turkey
| | - Rengül Çetin-Atalay
- Cancer Systems Biology Laboratory, Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara Turkey
| |
Collapse
|
13
|
Singh CK, George J, Chhabra G, Nihal M, Chang H, Ahmad N. Genetic Manipulation of Sirtuin 3 Causes Alterations of Key Metabolic Regulators in Melanoma. Front Oncol 2021; 11:676077. [PMID: 33937086 PMCID: PMC8085490 DOI: 10.3389/fonc.2021.676077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/29/2021] [Indexed: 12/29/2022] Open
Abstract
The mitochondrial sirtuin SIRT3 plays key roles in cellular metabolism and energy production, which makes it an obvious target for the management of cancer, including melanoma. Previously, we have demonstrated that SIRT3 was constitutively upregulated in human melanoma and its inhibition resulted in anti-proliferative effects in vitro in human melanoma cells and in vivo in human melanoma xenografts. In this study, we expanded our data employing knockdown and overexpression strategies in cell culture and mouse xenografts to further validate and establish the pro-proliferative function of SIRT3 in melanocytic cells, and its associated potential mechanisms, especially focusing on the metabolic regulation. We found that short-hairpin RNA (shRNA) mediated SIRT3 knockdown in G361 melanoma cells showed diminished tumorigenesis in immunodeficient Nu/Nu mice. Conversely, SIRT3 overexpressing Hs294T melanoma cells showed increased tumor growth. These effects were consistent with changes in markers of proliferation (PCNA), survival (Survivin) and angiogenesis (VEGF) in xenografted tissues. Further, in in vitro culture system, we determined the effect of SIRT3 knockdown on glucose metabolism in SK-MEL-2 cells, using a PCR array. SIRT3 knockdown caused alterations in a total of 37 genes involved in the regulation and enzymatic pathways of glucose (32 genes) and glycogen (5 genes) metabolism. Functions annotation of these identified genes, using the ingenuity pathway analysis (IPA), predicted cumulative actions of decreased cell viability/proliferation, tumor growth and reactive oxygen species (ROS), and increased apoptosis in response to SIRT3 knockdown. Further, IPA gene network analysis of SIRT3 modulated genes revealed the interactions among these genes in addition to several melanoma-associated genes. Sirtuin pathway was identified as one of the top canonical pathways showing the interaction of SIRT3 with metabolic regulatory genes along with other sirtuins. IPA analysis also predicted the inhibition of HIF1α, PKM, KDM8, PPARGC1A, mTOR, and activation of P53 and CLPP; the genes involved in major cancer/melanoma-associated signaling events. Collectively, these results suggest that SIRT3 inhibition affects cellular metabolism, to impart an anti-proliferative response against melanoma.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Jasmine George
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Minakshi Nihal
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Hao Chang
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, United States.,William S. Middleton VA Medical Center, Madison, WI, United States
| |
Collapse
|
14
|
Wei X, Shi J, Lin Q, Ma X, Pang Y, Mao H, Li R, Lu W, Wang Y, Liu P. Targeting ACLY Attenuates Tumor Growth and Acquired Cisplatin Resistance in Ovarian Cancer by Inhibiting the PI3K-AKT Pathway and Activating the AMPK-ROS Pathway. Front Oncol 2021; 11:642229. [PMID: 33816292 PMCID: PMC8011496 DOI: 10.3389/fonc.2021.642229] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/23/2021] [Indexed: 01/19/2023] Open
Abstract
Background: Ovarian cancer is the most lethal female genital malignancy. Although cisplatin is the first-line chemotherapy to treat ovarian cancer patients along with debulking surgeries, its efficacy is limited due to the high incidence of cisplatin resistance. ATP citrate lyase (ACLY) has been shown to be a key metabolic enzyme and is associated with poor prognosis in various cancers, including ovarian cancer. Nevertheless, no studies have probed the mechanistic relationship between ACLY and cisplatin resistance. Methods: Survival analysis was mainly carried out online. Bioinformatic analysis was performed in R/R studio. Proliferative activity was measured by MTT and colony formation assays. Cell cycle and apoptosis analysis were performed by flow cytometry. The acquired-cisplatin-resistant cell line A2780/CDDP was generated by exposing A2780 to cisplatin at gradually elevated concentrations. MTT assay was used to calculate IC50 values of cisplatin. A xenograft tumor assay was used test cell proliferation in vivo. Results: Higher expression of ACLY was found in ovarian cancer tissue and related to poor prognosis. Knockdown of ACLY in A2780, SKOV3, and HEY cells inhibited cell proliferation, caused cell-cycle arrest by modulating the P16–CDK4–CCND1 pathway, and induced apoptosis probably by inhibiting p-AKT activity. Bioinformatic analysis of the GSE15709 dataset revealed upregulation of ACLY and activation of PI3K–AKT pathway in cells with acquired cisplatin resistance, in line with observations on A2780/CDDP cells that we generated. Knockdown of ACLY alleviated cisplatin resistance, and works synergistically with cisplatin treatment to induce apoptosis in A2780/CDDP cells by inhibiting the PI3K–AKT pathway and activating AMPK–ROS pathway. The ACLY-specific inhibitor SB-204990 showed the same effect. In A2780/CDDP cells, AKT overexpression could attenuate cisplatin re-sensitization caused by ACLY knockdown. Conclusions: Knockdown of ACLY attenuated cisplatin resistance by inhibiting the PI3K–AKT pathway and activating the AMPK–ROS pathway. These findings suggest that a combination of ACLY inhibition and cisplatin might be an effective strategy for overcoming cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Xuan Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Juanjuan Shi
- Department of Gynecology and Obstetrics, Affiliated Tengzhou Center People's Hospital of Jining Medical University, Tengzhou, China
| | - Qianhan Lin
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoxue Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Hongluan Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Rui Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Lu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecology Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.,Shandong Engineering Laboratory for Urogynecology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
15
|
Gu L, Zhu Y, Lin X, Lu B, Zhou X, Zhou F, Zhao Q, Prochownik EV, Li Y. The IKKβ-USP30-ACLY Axis Controls Lipogenesis and Tumorigenesis. Hepatology 2021; 73:160-174. [PMID: 32221968 DOI: 10.1002/hep.31249] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 01/24/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death that develops as a consequence of obesity, cirrhosis, and chronic hepatitis. However, the pathways along which these changes occur remain incompletely understood. APPROACH AND RESULTS In this study, we show that the deubiquitinase USP30 is abundant in HCCs that arise in mice maintained on high-fat diets. IKKβ phosphorylated and stabilized USP30, which promoted USP30 to deubiquitinate ATP citrate lyase (ACLY) and fatty acid synthase (FASN). IKKβ also directly phosphorylated ACLY and facilitated the interaction between USP30 and ACLY and the latter's deubiquitination. In HCCs arising in DEN/CCl4 -treated mice, USP30 deletion attenuated lipogenesis, inflammation, and tumorigenesis regardless of diet. The combination of ACLY inhibitor and programmed death ligand 1 antibody largely suppressed chemical-induced hepatocarcinogenesis. The IKKβ-USP30-ACLY axis was also found to be up-regulated in human HCCs. CONCLUSIONS This study identifies an IKKβ-USP30-ACLY axis that plays an essential and wide-spread role in tumor metabolism and may be a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Li Gu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yahui Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xi Lin
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Bingjun Lu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xinyi Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Feng Zhou
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, China.,Hubei Clinical Center and Key Laboratory for Intestinal and Colorectal Diseases, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University School of Medicine, Wuhan, China.,Hubei Clinical Center and Key Laboratory for Intestinal and Colorectal Diseases, Wuhan, China
| | - Edward V Prochownik
- Division of Hematology/Oncology, Children's Hospital of Pittsburgh of UPMC, The Department of Microbiology and Molecular Genetics, The Pittsburgh Liver Research Center and The Hillman Cancer Center of UPMC, The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Ismail A, Doghish AS, E M Elsadek B, Salama SA, Mariee AD. Hydroxycitric acid potentiates the cytotoxic effect of tamoxifen in MCF-7 breast cancer cells through inhibition of ATP citrate lyase. Steroids 2020; 160:108656. [PMID: 32439410 DOI: 10.1016/j.steroids.2020.108656] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/17/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
Hydroxycitric acid (HCA), a dietary-derived weight loss supplement, competitively inhibits ATP citrate lyase (ACLY). Tamoxifen (TAM) is the most frequently used therapy for estrogen receptor (ER)-positive breast cancer patients, but its application was restricted due to efficacy related issues. Lipid metabolic reprogramming plays a key role in cancer progression and response to treatment. This study will test the hypothesis that targeting lipid metabolic enzymes could enhance TAM effect against breast cancer cells. MCF-7 ER-positive breast cancer cell line was used, and the cytotoxic effect of TAM treatment, alone and in combination with HCA was evaluated. Flowcytometric analysis of apoptosis following TAM and/or HCA treatment was additionally performed. Besides, the effects of TAM and/or HCA on ACLY, acetyl CoA carboxylase alpha (ACC-α) and fatty acid synthase (FAS) expression were investigated. Likewise, expression of ER-α protein through TAM and/or HCA treatment was examined. Cell contents of cholesterol and triglyceride were quantified. Treatment with TAM or HCA significantly reduced cell viability in a concentration-dependent manner whereas co-treatment synergistically reduced cell viability, promoted apoptosis, and decreased the expression of ACLY, ACC-α, and FAS. Intracellular triglyceride and cholesterol were accumulated in response to treatment with TAM and/or HCA. Moreover, either solitary TAM or TAM/ HCA co-treatment increased ER-α protein levels non significantly. Our results revealed that TAM effects on breast cancer are mediated, in part, through the regulation of key genes involved in lipid metabolism. Accordingly, inhibition of ACLY by HCA might be beneficial to enhance the therapeutic index of TAM against breast cancer.
Collapse
Affiliation(s)
- Ahmed Ismail
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, P.O. Box 11231, Nasr City, Cairo, Egypt.
| | - Ahmed S Doghish
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, P.O. Box 11231, Nasr City, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Bakheet E M Elsadek
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, Assuit Branch, P.O. Box 71524, Assuit, Egypt
| | - Salama A Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, P.O. Box 11231, Nasr City, Cairo, Egypt
| | - Amr D Mariee
- Biochemistry Department, Faculty of Pharmacy, Al-Azhar University, P.O. Box 11231, Nasr City, Cairo, Egypt
| |
Collapse
|
17
|
Targeting immunometabolism as an anti-inflammatory strategy. Cell Res 2020; 30:300-314. [PMID: 32132672 PMCID: PMC7118080 DOI: 10.1038/s41422-020-0291-z] [Citation(s) in RCA: 283] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
The growing field of immunometabolism has taught us how metabolic cellular reactions and processes not only provide a means to generate ATP and biosynthetic precursors, but are also a way of controlling immunity and inflammation. Metabolic reprogramming of immune cells is essential for both inflammatory as well as anti-inflammatory responses. Four anti-inflammatory therapies, DMF, Metformin, Methotrexate and Rapamycin all work by affecting metabolism and/or regulating or mimicking endogenous metabolites with anti-inflammatory effects. Evidence is emerging for the targeting of specific metabolic events as a strategy to limit inflammation in different contexts. Here we discuss these recent developments and speculate on the prospect of targeting immunometabolism in the effort to develop novel anti-inflammatory therapeutics. As accumulating evidence for roles of an intricate and elaborate network of metabolic processes, including lipid, amino acid and nucleotide metabolism provides key focal points for developing new therapies, we here turn our attention to glycolysis and the TCA cycle to provide examples of how metabolic intermediates and enzymes can provide potential novel therapeutic targets.
Collapse
|
18
|
The vital role of ATP citrate lyase in chronic diseases. J Mol Med (Berl) 2019; 98:71-95. [PMID: 31858156 DOI: 10.1007/s00109-019-01863-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
Chronic or non-communicable diseases are the leading cause of death worldwide; they usually result in long-term illnesses and demand long-term care. Despite advances in molecular therapeutics, specific biomarkers and targets for the treatment of these diseases are required. The dysregulation of de novo lipogenesis has been found to play an essential role in cell metabolism and is associated with the development and progression of many chronic diseases; this confirms the link between obesity and various chronic diseases. The main enzyme in this pathway-ATP-citrate lyase (ACLY), a lipogenic enzyme-catalyzes the critical reaction linking cellular glucose catabolism and lipogenesis. Increasing lines of evidence suggest that the modulation of ACLY expression correlates with the development and progressions of various chronic diseases such as neurodegenerative diseases, cardiovascular diseases, diabetes, obesity, inflammation, and cancer. Recent studies suggest that the inhibition of ACLY activity modulates the glycolysis and lipogenesis processes and stimulates normal physiological functions. This comprehensive review aimed to critically evaluate the role of ACLY in the development and progression of different diseases and the effects of its downregulation in the prevention and treatment of these diseases.
Collapse
|
19
|
Naeini MB, Momtazi AA, Jaafari MR, Johnston TP, Barreto G, Banach M, Sahebkar A. Antitumor effects of curcumin: A lipid perspective. J Cell Physiol 2019; 234:14743-14758. [PMID: 30741424 DOI: 10.1002/jcp.28262] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/06/2019] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Lipid metabolism plays an important role in cancer development due to the necessities of rapidly dividing cells to increase structural, energetic, and biosynthetic demands for cell proliferation. Basically, obesity, type 2 diabetes, and other related diseases, and cancer are associated with a common hyperactivated "lipogenic state." Recent evidence suggests that metabolic reprogramming and overproduction of enzymes involved in the synthesis of fatty acids are the new hallmarks of cancer, which occur in an early phase of tumorigenesis. As the first evidence to confirm dysregulated lipid metabolism in cancer cells, the overexpression of fatty acid synthase (FAS) was observed in breast cancer patients and demonstrated the role of FAS in cancer. Other enzymes of fatty acid synthesis have recently been found to be dysregulated in cancer, including ATP-dependent citrate lyase and acetyl-CoA carboxylase, which further underscores the connection of these metabolic pathways with cancer cell survival and proliferation. The degree of overexpression of lipogenic enzymes and elevated lipid utilization in tumors is closely associated with cancer progression. The question that arises is whether the progression of cancer can be suppressed, or at least decelerated, by modulating gene expression related to fatty acid metabolism. Curcumin, due to its effects on the regulation of lipogenic enzymes, might be able to suppress, or even cause regression of tumor growth. This review discusses recent evidence concerning the important role of lipogenic enzymes in the metabolism of cancer cells and whether the inhibitory effects of curcumin on lipogenic enzymes is therapeutically efficacious.
Collapse
Affiliation(s)
- Mehri Bemani Naeini
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri
| | - George Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Maciej Banach
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Gao J, Wang Y, Xu G, Wei J, Chang K, Tian X, Liu M, Yan X, Huo M, Song G. Selenium attenuates apoptosis and p-AMPK expressions in fluoride-induced NRK-52E cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:15685-15697. [PMID: 30949948 DOI: 10.1007/s11356-019-04855-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 03/13/2019] [Indexed: 06/09/2023]
Abstract
Fluoride is widely distributed in the environment, and excessive fluoride intake can induce cytotoxicity, DNA damage, and cell cycle changes in many tissues and organs, including the kidney. Accumulating evidence demonstrates that selenium (Se) administration ameliorates sodium fluoride (NaF)-induced kidney damage. However, the potentially beneficial effects of Se against NaF-induced cytotoxicity of the kidney and the underlying molecular mechanisms of this protection are not fully understood. At present, in this study, the normal rat kidney cell (NRK-52E) was used to investigate the potentially protective mechanism of Se against NaF-induced apoptosis, by using the methods of pathology, colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry, and Western blot. The experiment was designed with a control group, two NaF-treated groups (NaF, 5, 20 mg/L), two sodium selenite-treated groups (Na2SeO3, 17.1, 34.2 μg/L), and four Se + NaF-treated groups (Na2SeO3, 17.1, 34.2 μg/L; NaF, 5, 20 mg/L). The results indicate that selenium can attenuate apoptosis and AMPK phosphorylation in the NRK-52E cell induced with fluoride. These results imply that selenium is capable to modulate fluoride-induced NRK-52E cell apoptosis via regulating the expression levels of the proteins involved in mitochondrial pathway and changes in p-AMPK expressions may also be a key process in preventing fluorosis.
Collapse
Affiliation(s)
- Jiping Gao
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Yu Wang
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Guoqiang Xu
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Jianing Wei
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Kai Chang
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Xiaolin Tian
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Maolin Liu
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China
| | - Xiaoyan Yan
- School of Public Health, Shanxi Medical University, Shanxi, 030001, China
| | - Meijun Huo
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - Guohua Song
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Human Disease Animal Model, Shanxi Medical University, Road Xinjian 56, Taiyuan, 030001, China.
| |
Collapse
|
21
|
Morciano P, Di Giorgio ML, Porrazzo A, Licursi V, Negri R, Rong Y, Cenci G. Depletion of ATP-Citrate Lyase (ATPCL) Affects Chromosome Integrity Without Altering Histone Acetylation in Drosophila Mitotic Cells. Front Physiol 2019; 10:383. [PMID: 31019471 PMCID: PMC6458238 DOI: 10.3389/fphys.2019.00383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
The Citrate Lyase (ACL) is the main cytosolic enzyme that converts the citrate exported from mitochondria by the SLC25A1 carrier in Acetyl Coenzyme A (acetyl-CoA) and oxaloacetate. Acetyl-CoA is a high-energy intermediate common to a large number of metabolic processes including protein acetylation reactions. This renders ACL a key regulator of histone acetylation levels and gene expression in diverse organisms including humans. We have found that depletion of ATPCL, the Drosophila ortholog of human ACL, reduced levels of Acetyl CoA but, unlike its human counterpart, does not affect global histone acetylation and gene expression. Nevertheless, reduced ATPCL levels caused evident, although moderate, mitotic chromosome breakage suggesting that this enzyme plays a partial role in chromosome stability. These defects did not increase upon X-ray irradiation, indicating that they are not dependent on an impairment of DNA repair. Interestingly, depletion of ATPCL drastically increased the frequency of chromosome breaks (CBs) associated to mutations in scheggia, which encodes the ortholog of the mitochondrial citrate carrier SLC25A1 that is also required for chromosome integrity and histone acetylation. Our results indicate that ATPCL has a dispensable role in histone acetylation and prevents massive chromosome fragmentation when citrate efflux is altered.
Collapse
Affiliation(s)
- Patrizia Morciano
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza - Università di Roma, Rome, Italy
| | - Maria Laura Di Giorgio
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza - Università di Roma, Rome, Italy
| | - Antonella Porrazzo
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza - Università di Roma, Rome, Italy.,Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Valerio Licursi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Rodolfo Negri
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza - Università di Roma, Rome, Italy.,Istituto di Biologia e Patologia Molecolari (IBPM) del CNR, Rome, Italy
| | - Yikang Rong
- School of Life Sciences, State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, China
| | - Giovanni Cenci
- Dipartimento di Biologia e Biotecnologie "Charles Darwin", Sapienza - Università di Roma, Rome, Italy.,Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
22
|
Tumour microenvironment and metabolic plasticity in cancer and cancer stem cells: Perspectives on metabolic and immune regulatory signatures in chemoresistant ovarian cancer stem cells. Semin Cancer Biol 2018; 53:265-281. [DOI: 10.1016/j.semcancer.2018.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
23
|
Tan W, Zhong Z, Carney RP, Men Y, Li J, Pan T, Wang Y. Deciphering the metabolic role of AMPK in cancer multi-drug resistance. Semin Cancer Biol 2018; 56:56-71. [PMID: 30261277 DOI: 10.1016/j.semcancer.2018.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 09/02/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023]
Abstract
Multi-drug resistance (MDR) is a curious bottleneck in cancer research and chemotherapy, whereby some cells rapidly adapt to the tumor microenvironment via a myriad of heterogeneous metabolic activities. Despite being a major impediment to treatment, there is a silver lining: control over metabolic regulation could be an effective approach to overcome or correct resistance pathways. In this critical review, we comprehensively and carefully curated and analyzed large networks of previously identified proteins associated with metabolic adaptation in MDR. We employed data and text mining to study and categorize more than 600 studies in PubMed, with particular focus on AMPK, a central and fundamental modulator in the energy metabolism network that has been specifically implicated in cancer MDR pathways. We have identified one protein set of metabolic adaptations with 137 members closely related to cancer MDR processes, and a second protein set with 165 members derived from AMPK-based networks, with 28 proteins found at the intersection between the two sets. Furthermore, according to genomics analysis of the cancer genome atlas (TCGA) provisional data, the highest alteration frequency (80.0%) of the genes encoding the intersected proteins (28 proteins), ranked three cancer types with quite remarkable significance across 166 studies. The hierarchical relationships of the entire identified gene and protein networks indicate broad correlations in AMPK-mediated metabolic regulation pathways, which we use decipher and depict the metabolic roles of AMPK and demonstrate the potential of metabolic control for therapeutic intervention in MDR.
Collapse
Affiliation(s)
- Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu province 730000, China; Micro-Nano Innovations (MiNI) Laboratory, Biomedical Engineering, University of California, Davis, CA 95616, United States
| | - Zhangfeng Zhong
- Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60202, United States; Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau SAR, 999078, China
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States
| | - Yongfan Men
- Micro-Nano Innovations (MiNI) Laboratory, Biomedical Engineering, University of California, Davis, CA 95616, United States
| | - Jiannan Li
- Micro-Nano Innovations (MiNI) Laboratory, Biomedical Engineering, University of California, Davis, CA 95616, United States
| | - Tingrui Pan
- Micro-Nano Innovations (MiNI) Laboratory, Biomedical Engineering, University of California, Davis, CA 95616, United States.
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau SAR, 999078, China.
| |
Collapse
|
24
|
ATP citrate lyase (ACLY) inhibitors: An anti-cancer strategy at the crossroads of glucose and lipid metabolism. Eur J Med Chem 2018; 157:1276-1291. [DOI: 10.1016/j.ejmech.2018.09.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/31/2018] [Accepted: 09/01/2018] [Indexed: 02/06/2023]
|
25
|
Yadav SK, Pandey A, Kumar L, Devi A, Kushwaha B, Vishvkarma R, Maikhuri JP, Rajender S, Gupta G. The thermo-sensitive gene expression signatures of spermatogenesis. Reprod Biol Endocrinol 2018; 16:56. [PMID: 29859541 PMCID: PMC5985054 DOI: 10.1186/s12958-018-0372-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Spermatogenesis in most mammals (including human and rat) occurs at ~ 3 °C lower than body temperature in a scrotum and fails rapidly at 37 °C inside the abdomen. The present study investigates the heat-sensitive transcriptome and miRNAs in the most vulnerable germ cells (spermatocytes and round spermatids) that are primarily targeted at elevated temperature in a bid to identify novel targets for contraception and/or infertility treatment. METHODS Testes of adult male rats subjected to surgical cryptorchidism were obtained at 0, 24, 72 and 120 h post-surgery, followed by isolation of primary spermatocytes and round spermatids and purification to > 90% purity using a combination of trypsin digestion, centrifugal elutriation and density gradient centrifugation techniques. RNA isolated from these cells was sequenced by massive parallel sequencing technique to identify the most-heat sensitive mRNAs and miRNAs. RESULTS Heat stress altered the expression of a large number of genes by ≥2.0 fold, out of which 594 genes (286↑; 308↓) showed alterations in spermatocytes and 154 genes (105↑; 49↓) showed alterations in spermatids throughout the duration of experiment. 62 heat-sensitive genes were common to both cell types. Similarly, 66 and 60 heat-sensitive miRNAs in spermatocytes and spermatids, respectively, were affected by ≥1.5 fold, out of which 6 were common to both the cell types. CONCLUSION The study has identified Acly, selV, SLC16A7(MCT-2), Txnrd1 and Prkar2B as potential heat sensitive targets in germ cells, which may be tightly regulated by heat sensitive miRNAs rno-miR-22-3P, rno-miR-22-5P, rno-miR-129-5P, rno-miR-3560, rno-miR-3560 and rno-miR-466c-5P.
Collapse
Affiliation(s)
- Santosh K. Yadav
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
| | - Aastha Pandey
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
| | - Lokesh Kumar
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
| | - Archana Devi
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
- grid.469887.cAcademy of Scientific and Innovative Research (AcSIR), New Delhi, 110001 India
| | - Bhavana Kushwaha
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
- grid.469887.cAcademy of Scientific and Innovative Research (AcSIR), New Delhi, 110001 India
| | - Rahul Vishvkarma
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
| | - Jagdamba P. Maikhuri
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
| | - Singh Rajender
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
- grid.469887.cAcademy of Scientific and Innovative Research (AcSIR), New Delhi, 110001 India
| | - Gopal Gupta
- 0000 0004 0506 6543grid.418363.bDivision of Endocrinology, CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031 India
- grid.469887.cAcademy of Scientific and Innovative Research (AcSIR), New Delhi, 110001 India
| |
Collapse
|
26
|
Xin M, Qiao Z, Li J, Liu J, Song S, Zhao X, Miao P, Tang T, Wang L, Liu W, Yang X, Dai K, Huang G. miR-22 inhibits tumor growth and metastasis by targeting ATP citrate lyase: evidence in osteosarcoma, prostate cancer, cervical cancer and lung cancer. Oncotarget 2018; 7:44252-44265. [PMID: 27317765 PMCID: PMC5190093 DOI: 10.18632/oncotarget.10020] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 05/11/2016] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding small RNAs that function as negative regulators of gene expression involving in the tumor biology. ATP citrate lyase (ACLY), a key enzyme initiating de novo lipid synthesis, has been found to be upregulated in cancer cells, and its inhibition causes suppressive effects in a variety of tumors. At present, although several ACLY inhibitors have been reported, the potential role of miRNAs in interfering ACLY still needs further clarification. Herein, four different types of tumor cells including osteosarcoma, prostate, cervical and lung cancers were adopted in our study, and we have demonstrated that miR-22 directly downregulated ACLY. Moreover, miR-22 was proved to attenuate cancer cell proliferation and invasion, as well as promote cell apoptosis via inhibiting ACLY. Additionally, we confirmed the higher ACLY protein levels and the lower miR-22 expressions in hundreds of clinical samples of the four primary tumors, and a negative correlation relationship between ACLY and miR-22 was clarified. Finally, in the four animal models, we found that along with the loss of the ACLY expression, the miR-22-treated mice developed rather smaller tumors, less probabilities of distant metastasis, and fairly longer survivals. De novo lipogenesis suppression triggered by miR-22-ACLY axis may contribute to the inhibition of tumor growth and metastasis. These findings provide unequivocal proofs that miR-22 is responsible for the posttranscriptional regulation of ACLY, which yields promising therapeutic effects in osteosarcoma, prostate, cervical and lung cancers.
Collapse
Affiliation(s)
- Mei Xin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhiguang Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jing Li
- Bone and Joint Research Center, The First Affiliated Hospital, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710061, China.,Department of Orthopaedics, Alpert Medical School/Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shaoli Song
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ping Miao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Lei Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Weichun Liu
- Department of Gynecology and Obstetrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaodi Yang
- Department of Anesthesiology, Zhongshan Hospital, School of Medicine, Fudan University, Shanghai 200032, China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China.,The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200031, China
| |
Collapse
|
27
|
Williams NC, O'Neill LAJ. A Role for the Krebs Cycle Intermediate Citrate in Metabolic Reprogramming in Innate Immunity and Inflammation. Front Immunol 2018; 9:141. [PMID: 29459863 PMCID: PMC5807345 DOI: 10.3389/fimmu.2018.00141] [Citation(s) in RCA: 365] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/16/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolism in immune cells is no longer thought of as merely a process for adenosine triphosphate (ATP) production, biosynthesis, and catabolism. The reprogramming of metabolic pathways upon activation is also for the production of metabolites that can act as immune signaling molecules. Activated dendritic cells (DCs) and macrophages have an altered Krebs cycle, one consequence of which is the accumulation of both citrate and succinate. Citrate is exported from the mitochondria via the mitochondrial citrate- carrier. Cytosolic metabolism of citrate to acetyl-coenzyme A (acetyl-CoA) is important for both fatty-acid synthesis and protein acetylation, both of which have been linked to macrophage and DC activation. Citrate-derived itaconate has a direct antibacterial effect and also has been shown to act as an anti-inflammatory agent, inhibiting succinate dehydrogenase. These findings identify citrate as an important metabolite for macrophage and DC effector function.
Collapse
Affiliation(s)
- Niamh C Williams
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
28
|
Combination of Mitochondrial and Plasma Membrane Citrate Transporter Inhibitors Inhibits De Novo Lipogenesis Pathway and Triggers Apoptosis in Hepatocellular Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3683026. [PMID: 29546056 PMCID: PMC5818947 DOI: 10.1155/2018/3683026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/23/2017] [Accepted: 12/03/2017] [Indexed: 12/27/2022]
Abstract
Increased expression levels of both mitochondrial citrate transporter (CTP) and plasma membrane citrate transporter (PMCT) proteins have been found in various cancers. The transported citrates by these two transporter proteins provide acetyl-CoA precursors for the de novo lipogenesis (DNL) pathway to support a high rate of cancer cell viability and development. Inhibition of the DNL pathway promotes cancer cell apoptosis without apparent cytotoxic to normal cells, leading to the representation of selective and powerful targets for cancer therapy. The present study demonstrates that treatments with CTP inhibitor (CTPi), PMCT inhibitor (PMCTi), and the combination of CTPi and PMCTi resulted in decreased cell viability in two hepatocellular carcinoma cell lines (HepG2 and HuH-7). Treatment with citrate transporter inhibitors caused a greater cytotoxic effect in HepG2 cells than in HuH-7 cells. A lower concentration of combined CTPi and PMCTi promotes cytotoxic effect compared with either of a single compound. An increased cell apoptosis and an induced cell cycle arrest in both cell lines were reported after administration of the combined inhibitors. A combination treatment exhibits an enhanced apoptosis through decreased intracellular citrate levels, which consequently cause inhibition of fatty acid production in HepG2 cells. Apoptosis induction through the mitochondrial-dependent pathway was found as a consequence of suppressed carnitine palmitoyl transferase-1 (CPT-1) activity and enhanced ROS generation by combined CTPi and PMCTi treatment. We showed that accumulation of malonyl-CoA did not correlate with decreasing CPT-1 activity. The present study showed that elevated ROS levels served as an inhibition on Bcl-2 activity that is at least in part responsible for apoptosis. Moreover, inhibition of the citrate transporter is selectively cytotoxic to HepG2 cells but not in primary human hepatocytes, supporting citrate-mediating fatty acid synthesis as a promising cancer therapy.
Collapse
|
29
|
He Y, Gao M, Cao Y, Tang H, Liu S, Tao Y. Nuclear localization of metabolic enzymes in immunity and metastasis. Biochim Biophys Acta Rev Cancer 2017; 1868:359-371. [PMID: 28757126 DOI: 10.1016/j.bbcan.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
Metabolism is essential to all living organisms that provide cells with energy, regulators, building blocks, enzyme cofactors and signaling molecules, and is in tune with nutritional conditions and the function of cells to make the appropriate developmental decisions or maintain homeostasis. As a fundamental biological process, metabolism state affects the production of multiple metabolites and the activation of various enzymes that participate in regulating gene expression, cell apoptosis, cancer progression and immunoreactions. Previous studies generally focus on the function played by the metabolic enzymes in the cytoplasm and mitochondrion. In this review, we conclude the role of them in the nucleus and their implications for cancer progression, immunity and metastasis.
Collapse
Affiliation(s)
- Yuchen He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Menghui Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiqu Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
30
|
Migita T, Takayama KI, Urano T, Obinata D, Ikeda K, Soga T, Takahashi S, Inoue S. ACSL3 promotes intratumoral steroidogenesis in prostate cancer cells. Cancer Sci 2017; 108:2011-2021. [PMID: 28771887 PMCID: PMC5623750 DOI: 10.1111/cas.13339] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/06/2017] [Accepted: 07/30/2017] [Indexed: 01/12/2023] Open
Abstract
Long‐chain acyl‐coenzyme A (CoA) synthetase 3 (ACSL3) is an androgen‐responsive gene involved in the generation of fatty acyl‐CoA esters. ACSL3 is expressed in both androgen‐sensitive and castration‐resistant prostate cancer (CRPC). However, its role in prostate cancer remains elusive. We overexpressed ACSL3 in androgen‐dependent LNCaP cells and examined the downstream effectors of ACSL3. Furthermore, we examined the role of ACSL3 in the androgen metabolism of prostate cancer. ACSL3 overexpression led to upregulation of several genes such as aldo‐keto reductase 1C3 (AKR1C3) involved in steroidogenesis, which utilizes adrenal androgen dehydroepiandrosterone sulfate (DHEAS) as substrate, and downregulated androgen‐inactivating enzyme UDP‐glucuronosyltransferase 2 (UGT2B). Exposure to DHEAS significantly increased testosterone levels and cell proliferative response in ACSL3‐overexpressing cells when compared to that in control cells. A public database showed that ACSL3 level was higher in CRPC than in hormone‐sensitive prostate cancer. CRPC cells showed an increased expression of ACSL3 and an expression pattern of AKR1C3 and UGT2B similar to ACSL3‐overexpressing cells. DHEAS stimulation significantly promoted the proliferation of CRPC cells when compared to that of LNCaP cells. These findings suggest that ACSL3 contributes to the growth of CRPC through intratumoral steroidogenesis (i.e. promoting androgen synthesis from DHEAS and preventing the catabolism of active androgens).
Collapse
Affiliation(s)
- Toshiro Migita
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken-Ichi Takayama
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomohiko Urano
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Daisuke Obinata
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Kazutaka Ikeda
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan.,RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Inoue
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
31
|
Wang D, Yin L, Wei J, Yang Z, Jiang G. ATP citrate lyase is increased in human breast cancer, depletion of which promotes apoptosis. Tumour Biol 2017; 39:1010428317698338. [PMID: 28443474 DOI: 10.1177/1010428317698338] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is a malignant tumor that is harmful to women’s health around the world. Investigating the biological mechanism is, therefore, of pivotal importance to improve patients’ prognoses. Compared to non-neoplastic tissues, enhanced glucose and lipid metabolism is one of the most common properties of malignant breast cancer. Adenosine triphosphate (ATP) citrate lyase is a key enzyme linking aerobic glycolysis and fatty acid synthesis and is of high biological and prognostic significance in breast cancer. In our clinical study, fresh clinical tissues were used to analyze ATP citrate lyase expression by western blotting, and paraffin archived samples from 62 breast cancer patients were used to analyze ATP citrate lyase expression by immunohistochemistry. In the cellular study, following small interfering RNA–mediated inhibition of ATP citrate lyase in MCF-7 cells, cell viability and apoptosis were measured using the Cell Counting Kit-8 and flow cytometry, respectively. Breast cancer tissues showed strong expression of ATP citrate lyase, whereas adjacent normal tissues showed weak expression. Silencing of endogenous ATP citrate lyase expression by small interfering RNA in MCF-7 cells suppressed cell viability and increased cell apoptosis. Collectively, our study revealed that expression of ATP citrate lyase was significantly increased in breast cancer tissue compared with normal tissue. In addition, we found that depletion of ATP citrate lyase suppressed tumor growth, which suggests that ATP citrate lyase–related inhibitors might be potential therapeutic approaches for breast cancer.
Collapse
Affiliation(s)
- Diyu Wang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Yin
- Department of General Surgery, Suzhou Wuzhong People’s Hospital, Suzhou, China
| | - Jinrong Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhixue Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guoqin Jiang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
32
|
Guerram M, Jiang ZZ, Yousef BA, Hamdi AM, Hassan HM, Yuan ZQ, Luo HW, Zhu X, Zhang LY. The potential utility of acetyltanshinone IIA in the treatment of HER2-overexpressed breast cancer: Induction of cancer cell death by targeting apoptotic and metabolic signaling pathways. Oncotarget 2016; 6:21865-77. [PMID: 26068969 PMCID: PMC4673132 DOI: 10.18632/oncotarget.4156] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/14/2015] [Indexed: 01/21/2023] Open
Abstract
Increased lipogenesis and protein synthesis is a hallmark of cancer cell proliferation, survival, and metastatic progression and is under intense investigation as a potential antineoplastic target. Acetyltanshinone IIA (ATA) is a compound that was obtained from chemical modifications of tanshinone IIA (TIIA), a potent anticancer agent extracted from the dried roots of the Chinese herbal medicine Salvia miltiorrhiza Bunge. A previous investigation indicated that ATA is more effective in inhibiting the growth of breast cancer especially cells with HER2 overexpression. However, the molecular mechanism(s) mediating this cytotoxic effect on HER2-positive breast cancer remained undefined. Studies described here report that ATA induced G1/S phase arrest and apoptosis in the HER2-positive MDA-MB-453, SK-BR-3, and BT-474 breast cancer cell lines. Mechanistic investigations revealed that the ATA-induced apoptosis effect is associated with remarkably down-regulation of receptor tyrosine kinases (RTKs) EGFR/HER2 and inhibition of their downstream pro-survival signaling pathways. Interestingly, ATA was found to trigger oxidative and endoplasmic reticulum (ER) stresses and to activate AMP activated protein kinase (AMPK) leading to inactivation of key enzymes involved in lipid and protein biogenesis. Intraperitoneal administration of ATA significantly inhibited the growth of MDA-MB-453 xenografts in athymic mice without causing weight loss and any other side effects. Additionally, transwell migration, invasion, and wound healing assays revealed that ATA could suppress tumor angiogenesis in vitro. Taken together, our data suggest that ATA may have broad utility in the treatment of HER2-overexpressed breast cancers.
Collapse
Affiliation(s)
- Mounia Guerram
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Bashir Alsiddig Yousef
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Aida Mejda Hamdi
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Hozeifa Mohamed Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zi-Qiao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Hou-Wei Luo
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xiong Zhu
- Medical and Chemical Institute, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
33
|
Feng YH, Chen WY, Kuo YH, Tung CL, Tsao CJ, Shiau AL, Wu CL. Elovl6 is a poor prognostic predictor in breast cancer. Oncol Lett 2016; 12:207-212. [PMID: 27347126 DOI: 10.3892/ol.2016.4587] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 04/29/2016] [Indexed: 12/26/2022] Open
Abstract
Elongation of long chain fatty acids family member 6 (Elovl6) has been demonstrated to be involved in insulin resistance, obesity and lipogenesis. In addition, it has been reported that the protein is upregulated in human hepatocellular carcinoma and is implicated in nonalcoholic steatohepatitis-associated liver carcinogenesis. Excess body weight has been associated with an increased risk of postmenopausal breast cancer and poor prognosis. However, the connection between Elovl6 expression and outcome of breast cancer remains uncertain. Therefore, the present study used immunohistochemical analysis to investigate the expression of Elovl6 in breast cancer tissues from patients who had undergone curative mastectomy. Out of a total of 70 patients, 37.1% of patients exhibited positive Elovl6 expression in breast cancer tissue, whilst 62.9% were considered as negative. Positive Elov16 expression correlated with positive lymph node involvement and shorter recurrence-free survival. However, Elovl6 expression had no association with primary tumor size, lymph node metastasis, stage, grade, estrogen receptor, progesterone receptor, HER2 and age. Therefore, positive Elovl6 expression is a poor prognostic factor in patients with breast cancer that have previously undergone surgery, and may function as a potential therapeutic approach in the future, particularly in the scope of obesity related disease.
Collapse
Affiliation(s)
- Yin-Hsun Feng
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan R.O.C.; Department of Nursing, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan R.O.C
| | - Wei-Yu Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan R.O.C
| | - Yu-Hsuan Kuo
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan R.O.C
| | - Chao-Ling Tung
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan R.O.C
| | - Chao-Jung Tsao
- Department of Hematology and Oncology, Chi-Mei Medical Center, Tainan 73657, Taiwan R.O.C
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan R.O.C
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan R.O.C
| |
Collapse
|
34
|
Zhong ZF, Tan W, Qiang WW, Scofield VL, Tian K, Wang CM, Qiang WA, Wang YT. Furanodiene alters mitochondrial function in doxorubicin-resistant MCF-7 human breast cancer cells in an AMPK-dependent manner. MOLECULAR BIOSYSTEMS 2016; 12:1626-37. [DOI: 10.1039/c6mb00003g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Furanodiene is a bioactive sesquiterpene isolated from the spice-producing Curcuma wenyujin plant (Y. H. Chen and C. Ling) (C. wenyujin), which is a commonly prescribed herb used in clinical cancer therapy by modern practitioners of traditional Chinese medicine.
Collapse
Affiliation(s)
- Zhang-Feng Zhong
- Institute of Chinese Medical Sciences
- State Key Laboratory of Quality Research in Chinese Medicine
- University of Macau
- Avenida da Universidade
- Taipa
| | - Wen Tan
- School of Pharmacy
- Lanzhou University
- Lanzhou
- China
| | - William W. Qiang
- Institute of Chinese Medical Sciences
- State Key Laboratory of Quality Research in Chinese Medicine
- University of Macau
- Avenida da Universidade
- Taipa
| | - Virginia L. Scofield
- Department of Biomedical Sciences
- School of Medicine
- University of Texas Rio Grande Valley
- Edinburg
- USA
| | - Ke Tian
- School of Chinese Medicine
- Hong Kong Baptist University
- Hong Kong 999077
- China
- Division of Reproductive Science in Medicine
| | - Chun-Ming Wang
- Institute of Chinese Medical Sciences
- State Key Laboratory of Quality Research in Chinese Medicine
- University of Macau
- Avenida da Universidade
- Taipa
| | - Wen-An Qiang
- Division of Reproductive Science in Medicine
- Department of Obstetrics and Gynecology
- Feinberg School of Medicine at Northwestern University
- Chicago
- USA
| | - Yi-Tao Wang
- Institute of Chinese Medical Sciences
- State Key Laboratory of Quality Research in Chinese Medicine
- University of Macau
- Avenida da Universidade
- Taipa
| |
Collapse
|
35
|
Csanadi A, Kayser C, Donauer M, Gumpp V, Aumann K, Rawluk J, Prasse A, zur Hausen A, Wiesemann S, Werner M, Kayser G. Prognostic Value of Malic Enzyme and ATP-Citrate Lyase in Non-Small Cell Lung Cancer of the Young and the Elderly. PLoS One 2015; 10:e0126357. [PMID: 25962060 PMCID: PMC4427316 DOI: 10.1371/journal.pone.0126357] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/01/2015] [Indexed: 01/29/2023] Open
Abstract
Background Lung cancer is the leading cause of death among malignancies worldwide. Understanding its biology is therefore of pivotal importance to improve patient’s prognosis. In contrast to non-neoplastic tissues, cancer cells utilize glucose mainly for production of basic cellular modules ‘(i.e. nucleotides, aminoacids, fatty acids). In cancer, Malic enzyme (ME) and ATP-citrate lyase (ACLY) are key enzymes linking aerobic glycolysis and fatty acid synthesis and may therefore be of biological and prognostic significance in non-small cell lung cancer (NSCLC). Material and Methods ME and ACLY expression was analyzed in 258 NSCLC in correlation with clinico-pathological parameters including patient’s survival. Results Though, overall expression of both enzymes correlated positively, ACLY was associated with local tumor stage, whereas ME correlated with occurrence of mediastinal lymph node metastases. Young patients overexpressing ACLY and/or ME had a significantly longer overall survival. This proved to be an independent prognostic factor. This contrasts older NSCLC patients, in whom overexpression of ACLY and/or ME appears to predict the opposite. Conclusion In NSCLC, ME and ACLY show different enzyme expressions relating to local and mediastinal spread. Most important, we detected an inverse prognostic impact of ACLY and/or ME overexpression in young and elderly patients. It can therefore be expected, that treatment of NSCLC especially, if targeting metabolic pathways, requires different strategies in different age groups.
Collapse
Affiliation(s)
- Agnes Csanadi
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
| | - Claudia Kayser
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
| | - Marcel Donauer
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
| | - Vera Gumpp
- Clinical Cancer Registry, Comprehensive Cancer Center Freiburg, University Medical Center Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | - Konrad Aumann
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
| | - Justyna Rawluk
- Department of Hematology and Oncology, University Medical Center Freiburg, Hugstetter Strassse 55, D-79106 Freiburg, Germany
| | - Antje Prasse
- Department of Pneumonology, Hannover Medical School, Carl-Neuberg Strasse 1, D-30625 Hannover, Germany
| | - Axel zur Hausen
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sebastian Wiesemann
- Department of Thoracic Surgery, University Medical Center Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | - Martin Werner
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
| | - Gian Kayser
- Department of Pathology, Institute of Surgical Pathology, University Medical Center Freiburg, Breisacher Strasse 115a, D-79106 Freiburg, Germany
- * E-mail:
| |
Collapse
|
36
|
Kawaguchi T, Hayakawa M, Koga H, Torimura T. Effects of fucoidan on proliferation, AMP-activated protein kinase, and downstream metabolism- and cell cycle-associated molecules in poorly differentiated human hepatoma HLF cells. Int J Oncol 2015; 46:2216-22. [PMID: 25776104 DOI: 10.3892/ijo.2015.2928] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 02/27/2015] [Indexed: 11/05/2022] Open
Abstract
Survival rates are low in patients with poorly differentiated hepatocellular carcinoma (HCC). Fucoidan, a sulfated polysaccharide derived from brown seaweed, has anticancer activity; however, the effects of fucoidan on poorly differentiated HCC remain unclear. In this study, we investigated the effects of fucoidan on AMP-activated protein kinase (AMPK), a proliferation regulator, and its downstream metabolism- and cell cycle-related molecules in a poorly differentiated human hepatoma HLF cell line. HLF cells were treated with fucoidan (10, 50, or 100 µg/ml; n=4) or phosphate buffered saline (control; n=4) for 96 h. Proliferation was evaluated by counting cells every 24 h. AMPK, TSC2, mTOR, GSK3β, acetyl-CoA carboxylase (ACC), ATP-citrate lyase, p53, cyclin D1, cyclin-dependent kinase (CDK) 4, and CDK6 expression and/or phosphorylation were examined by immunoblotting 24 h after treatment with 100 µg/ml fucoidan. Cell cycle progression was analyzed by fluorescence-activated cell sorter 48 h after treatment. Treatment with 50 or 100 µg/ml fucoidan significantly and dose- and time-dependently suppressed HLF cell proliferation (P<0.0001). Fucoidan induced AMPK phosphorylation on Ser172 24 h after treatment. Although no differences were seen in expression and phosphorylation levels of TSC2, mTOR, GSK3β, ATP-citrate lyase, and p53 between the control and fucoidan-treated HLF cells, fucoidan induced ACC phosphorylation on Ser79. Moreover, fucoidan decreased cyclin D1, CDK4 and CDK6 expression 24 h after treatment. Furthermore, HLF cells were arrested in the G1/S phase 48 h after fucoidan treatment. We demonstrated that fucoidan suppressed HLF cell proliferation with AMPK phosphorylation. We showed that fucoidan phosphorylated ACC and downregulated cyclin D1, CDK4 and CDK6 expression. Our findings suggest that fucoidan inhibits proliferation through AMPK-associated suppression of fatty acid synthesis and G1/S transition in HLF cells.
Collapse
Affiliation(s)
- Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masako Hayakawa
- Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
37
|
Zong H, Zhang Y, You Y, Cai T, Wang Y. RETRACTED ARTICLE: Decreased Warburg effect induced by ATP citrate lyase suppression inhibits tumor growth in pancreatic cancer. Med Oncol 2015; 32:85. [DOI: 10.1007/s12032-015-0540-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/13/2015] [Indexed: 12/25/2022]
|
38
|
Xie S, Zhou F, Wang J, Cao H, Chen Y, Liu X, Zhang Z, Dai J, He X. Functional polymorphisms of ATP citrate lyase gene predicts clinical outcome of patients with advanced colorectal cancer. World J Surg Oncol 2015; 13:42. [PMID: 25890184 PMCID: PMC4359538 DOI: 10.1186/s12957-015-0440-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/07/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Previous studies have demonstrated that ATP citrate lyase (ACLY) plays an important role in the development of many cancers. Our current study aims to assess the effects of functional single nucleotide polymorphisms (SNPs) in ACLY gene on recurrence and survival of colorectal cancer (CRC) patients. METHODS A total of 697 resected Chinese CRC patients were included in this study. Two functional single nucleotide polymorphisms in ACLY gene were examined using the Sequenom iPLEX genotyping system. Multivariate Cox proportional hazards model and Kaplan-Meier curve were used for the prognosis analysis. RESULTS Multivariate Cox regression analysis showed that there was no significant association between SNPs in ACLY gene and the prognosis of total patient cohort. However, in patients with stage III + IV diseases, the two functional SNPs (rs2304497 and rs9912300) exhibited a significant association with the risks of death (HR = 0.47, 95% CI = 0.24-0.90 and HR = 0.59, 95% CI = 0.37-0.92, respectively) and recurrence (HR = 0.46, 95% CI = 0.24-0.86 and HR = 0.54, CI = 0.35-0.83, respectively). Kaplan-Meier analysis indicated that those CRC patients carrying heterozygous (WV) or homozygous variant (VV) genotypes in rs2304497 and rs9912300 had significantly better overall survival (OS) and recurrence-free survival (RFS). Moreover, we observed remarkable cumulative effects of these two SNPs on overall survival and recurrence-free survival (P for trend = 0.012 and 0.003, respectively). Compared with patients carrying zero unfavorable genotype, those carrying two unfavorable genotypes had a 2.24-fold and 2.33-fold increase of death and recurrence risks, respectively. CONCLUSIONS The SNPs in ACLY gene may serve as independent prognostic markers for patients with advanced stage CRC.
Collapse
Affiliation(s)
- Shuang Xie
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, 169 West Changle Street, Xi'an, 710032, China. .,State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Feng Zhou
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, 169 West Changle Street, Xi'an, 710032, China. .,Department of General Surgery, Huaihai Hospital, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China.
| | - Jiaojiao Wang
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Haiyan Cao
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yibing Chen
- State Key Laboratory of Cancer Biology, Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiaonan Liu
- Xijing Hospital of Digestive Disease, Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhaohui Zhang
- Department of General Surgery, Huaihai Hospital, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China.
| | - Jingyao Dai
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, 169 West Changle Street, Xi'an, 710032, China.
| |
Collapse
|
39
|
Gao Y, Islam MS, Tian J, Lui VWY, Xiao D. Inactivation of ATP citrate lyase by Cucurbitacin B: A bioactive compound from cucumber, inhibits prostate cancer growth. Cancer Lett 2014; 349:15-25. [PMID: 24690568 DOI: 10.1016/j.canlet.2014.03.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 01/03/2023]
Abstract
Prostate cancer, a leading cause of cancer-related deaths in males, is well recognized as having late disease on-set (mostly at age 60-70) and showing slow/latent disease development, and strategies to prevent cancer formation in late manhood may have significant health impacts. Cucurbitacin B (CuB) is a naturally occurring compound that is found abundantly in cucumbers and other vegetables, and it is known to exert anti-cancer activities (primarily via apoptosis-induction) in several human cancers. However, its chemopreventive potential for prostate cancer has not yet been investigated. Here, we reported that CuB significantly and specifically inhibited prostate cancer cell growth with low IC50 (~0.3 μM; PC-3 and LNCaP), accompanied by marked apoptosis (Caspase 3/7 activation, PARP cleavage, increase of Annexin V-Alexa Fluor 488 (Alexa488)+ cells and accumulation of Sub-G0/G1 population), whereas normal human prostate epithelial cells (PrEC) were CuB-insensitive. Using a chemopreventive model, pre-treatment of mice with CuB (2 weeks before PC-3 prostate cancer cell implantation) significantly reduced the rate of in vivo tumor-formation. A 79% reduction in tumor size (accompanied by marked in situ apoptosis) was observed in the CuB-treated group (with no noticeable toxicity) vs. controls at day 31. Strikingly, mechanistic investigations demonstrated that CuB drove dose-dependent inhibition of ATP citrate lyase phosphorylation (ACLY; an important enzyme for cancer metabolism) both in vitro and in the CuB-chemopreventive mouse model. Importantly, ACLY over-expression abrogated CuB's apoptotic effects in prostate cancer cells, confirming ACLY as a direct target of CuB. Thus, CuB harbors potent chemopreventive activity for prostate cancer, and we revealed a novel anti-tumor mechanism of CuB via inhibition of ACYL signaling in human cancer.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Mohammad Shyful Islam
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Jiang Tian
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Vivian Wai Yan Lui
- Department of Pharmacology and Pharmacy, Li Ka Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region
| | - Dong Xiao
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
40
|
Migita T, Okabe S, Ikeda K, Igarashi S, Sugawara S, Tomida A, Soga T, Taguchi R, Seimiya H. Inhibition of ATP citrate lyase induces triglyceride accumulation with altered fatty acid composition in cancer cells. Int J Cancer 2013; 135:37-47. [PMID: 24310723 DOI: 10.1002/ijc.28652] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/16/2013] [Accepted: 11/19/2013] [Indexed: 01/17/2023]
Abstract
De novo lipogenesis is activated in most cancers and several lipogenic enzymes have been implicated as therapeutic targets. Here, we demonstrate a novel function of the lipogenic enzyme, ATP citrate lyase (ACLY), in lipid metabolism in cancer cells. ACLY depletion by small interfering RNAs caused growth suppression and/or apoptosis in a subset of cancer cell lines. To investigate the effect of ACLY inhibition on lipid metabolism, metabolome and transcriptome analysis was performed. ACLY depletion blocks the fatty acid chain elongation from C16 to C18 in triglyceride (TG), but not in other lipid classes. Meanwhile, wild-type ACLY overexpression enhanced fatty acid elongation of TG, whereas an inactive mutant ACLY did not change it. ACLY depletion-mediated blockade of fatty acid elongation was coincident with downregulation of long-chain fatty acid elongase ELOVL6, which resides in endoplasmic reticulum (ER). Paradoxically, ACLY depletion-mediated growth suppression was associated with TG accumulation. ACLY depletion downregulated the expression of carnitine palmitoyltransferase 1A, which is a mitochondrial fatty acid transporter. Consistent with this finding, metabolome analysis revealed that ACLY positively regulates the carnitine system, which plays as an essential cofactor for fatty acid transport across mitochondrial membrane. AICAR, an activator of mitochondrial fatty acid oxidation (FAO), significantly reduced ACLY depletion-mediated TG accumulation. These data indicate that inhibition of ACLY might affect both fatty acid elongation in ER and FAO in mitochondria, thereby explaining the TG accumulation with altered fatty acid composition. This phenotype may be a hallmark of growth suppression mediated by ACLY inhibition.
Collapse
Affiliation(s)
- Toshiro Migita
- Division of Molecular Biotherapy Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhang J, Hochwald SN. The role of FAK in tumor metabolism and therapy. Pharmacol Ther 2013; 142:154-63. [PMID: 24333503 DOI: 10.1016/j.pharmthera.2013.12.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/08/2013] [Indexed: 02/06/2023]
Abstract
Focal adhesion kinase (FAK) plays a vital role in tumor cell proliferation, survival and migration. Altered metabolic pathways fuel rapid tumor growth by accelerating glucose, lipid and glutamine processing. Besides the mitogenic effects of FAK, evidence is accumulating supporting the association between hyper-activated FAK and aberrant metabolism in tumorigenesis. FAK can promote glucose consumption, lipogenesis, and glutamine dependency to promote cancer cell proliferation, motility, and survival. Clinical studies demonstrate that FAK-related alterations of tumor metabolism are associated with increased risk of developing solid tumors. Since FAK contributes to the malignant phenotype, small molecule inhibition of FAK-stimulated bioenergetic and biosynthetic processes can provide a novel approach for therapeutic intervention in tumor growth and invasion.
Collapse
Affiliation(s)
- Jianliang Zhang
- Department of Surgical Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Steven N Hochwald
- Department of Surgical Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States.
| |
Collapse
|