1
|
Chen J, Zhao D, Wang Y, Liu M, Zhang Y, Feng T, Xiao C, Song H, Miao R, Xu L, Chen H, Qiu X, Xu Y, Xu J, Cui Z, Wang W, Quan Y, Zhu Y, Huang C, Zheng SG, Zhao J, Zhu T, Sun L, Fan G. Lactylated Apolipoprotein C-II Induces Immunotherapy Resistance by Promoting Extracellular Lipolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406333. [PMID: 38981044 PMCID: PMC11481198 DOI: 10.1002/advs.202406333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Indexed: 07/11/2024]
Abstract
Mortality rates due to lung cancer are high worldwide. Although PD-1 and PD-L1 immune checkpoint inhibitors boost the survival of patients with non-small-cell lung cancer (NSCLC), resistance often arises. The Warburg Effect, which causes lactate build-up and potential lysine-lactylation (Kla), links immune dysfunction to tumor metabolism. The role of non-histone Kla in tumor immune microenvironment and immunotherapy remains to be clarified. Here, global lactylome profiling and metabolomic analyses of samples from patients with NSCLC is conducted. By combining multi-omics analysis with in vitro and in vivo validation, that intracellular lactate promotes extracellular lipolysis through lactyl-APOC2 is revealed. Mechanistically, lactate enhances APOC2 lactylation at K70, stabilizing it and resulting in FFA release, regulatory T cell accumulation, immunotherapy resistance, and metastasis. Moreover, the anti-APOC2K70-lac antibody that sensitized anti-PD-1 therapy in vivo is developed. This findings highlight the potential of anti lactyl-APOC2-K70 approach as a new combination therapy for sensitizing immunotherapeutic responses.
Collapse
Affiliation(s)
- Jian Chen
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Yupeng Wang
- Department of General Surgery, Shanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghai200011P. R. China
| | - Ming Liu
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Yuan Zhang
- Department of Gastrointestinal SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Tingting Feng
- Department of Clinical PharmacyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Chao Xiao
- Department of Gastrointestinal SurgeryShanghai East Hospital, School of MedicineTongji UniversityShanghai200040P. R. China
| | - Huan Song
- Department of Clinical Laboratory MedicineShanghai Pulmonary HospitalTongji University School of MedicineShanghai200433P. R. China
| | - Rui Miao
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Li Xu
- Department of Thoracic Surgery, Shanghai Pulmonary HospitalTongji University507 Zhengmin RoadShanghai200433P. R. China
| | - Hongwei Chen
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Xiaoying Qiu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Yi Xu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Jingxuan Xu
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty MannheimHeidelberg University68167MannheimGermany
| | - Zelin Cui
- Department of Laboratory MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Wei Wang
- Department of Breast‐thyroid SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Yanchun Quan
- Central LaboratoryLinyi People's HospitalShandong273300P. R. China
| | - Yifeng Zhu
- Department of Internal Medicine II, Klinikum rechts der IsarTechnical University of MunichIsmaninger Str. 2281675MunichGermany
| | - Chen Huang
- Department of Gastrointestinal SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620P. R. China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Songjiang Research InstituteShanghai Jiaotong University School of Medicine Affiliated Songjiang HospitalShanghai200080P. R. China
| | - Jian‐yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Ting Zhu
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Lianhui Sun
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Guangjian Fan
- Precision Research Center for Refractory Diseases, Institute for Clinical ResearchShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| |
Collapse
|
2
|
Yang T, Qiao S, Zhu X. High-dose radiation-resistant lung cancer cells stored many functional lipid drops through JAK2/p-STAT3/FASN pathway. J Cancer Res Clin Oncol 2023; 149:14169-14183. [PMID: 37553421 DOI: 10.1007/s00432-023-05106-1] [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: 04/21/2023] [Accepted: 06/30/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND The understanding of radiation resistance is still unclear. This study aims to explore the new mechanism of radiation resistance in lung cancer from the perspective of lipid metabolism. METHODS Oil red O was used to detect the amount of lipid droplets in high-dose radiation-resistant lung cancer cells (HDRR-LCCs) and the primary lung cancer cells. Western blot analysis was used to determine the protein expression levels of key molecules related to de novo fatty acid synthesis and fatty acid transport. Orlistat was used to inhibit the de novo fatty acid synthesis. The prediction of the transcriptional regulators of fatty acid synthetase (FASN) was analyzed by bioinformatics. AZD-1480 was used to inhibit the JAK2/STAT3 pathway to observe its effects on FASN and intracellular lipid droplets. The regulation of the transcription factor p-STAT3 on the FASN gene was verified by Chip-qPCR. Finally, we used the public data of lung cancer patients to analyze the correlation between FASN and LPL gene expression with the prognosis. RESULTS There were more lipid drops in the HDRR-LCCs than in the primary lung cancer cells. HDRR-LCCs preferred de novo synthesis of fatty acids, and high expression of LPL homodimers indicated a high intake of extracellular fatty acids. The expression of FASN was increased in HDRR-LCCs compared with the primary lung cancer cells in a radiation-dose-dependent way, while LPL homodimers did not show such a trend. The lipid droplets, cell proliferation, and radiation resistance were decreased in HDRR-LCCs after orlistat treatment. Lipid droplets were significantly reduced, and the protein expression of FASN also decreased when using AZD-1480 to inhibit the JAK2/STAT3 pathway. The Chip-qPCR showed that p-STAT3 was the upstream regulator which binds to the promoter region of FASN. Survival analysis showed that high expression of the FASN gene was associated with a poor prognosis in lung cancer patients who received radiotherapy. CONCLUSION Our studies discovered that lipids deposited in HDRR-LCCs were due to endogenous de novo fatty acids synthesis and exogenous lipids uptake. JAK2/p-TAT3/FASN could be used as promising targets for radiotherapy sensitization. Our study provided a new theoretical basis for studying the mechanism of radiation resistance in lung cancer.
Collapse
Affiliation(s)
- Ting Yang
- Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Simiao Qiao
- Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Xiaoxia Zhu
- Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
3
|
Morita T. Seeking an Important Role on Metabolomics—Effects of β-Estradiol on Lipoprotein Metabolism in Mammary Tumors. YAKUGAKU ZASSHI 2022; 142:1191-1199. [DOI: 10.1248/yakushi.22-00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tetsuo Morita
- Department of Biochemistry, Faculty and Graduate School of Pharmacy and Pharmaceutical Sciences, Fukuyama University
| |
Collapse
|
4
|
A Toolbox for Functional Analysis and the Systematic Identification of Diagnostic and Prognostic Gene Expression Signatures Combining Meta-Analysis and Machine Learning. Cancers (Basel) 2019; 11:cancers11101606. [PMID: 31640282 PMCID: PMC6827106 DOI: 10.3390/cancers11101606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023] Open
Abstract
The identification of biomarker signatures is important for cancer diagnosis and prognosis. However, the detection of clinical reliable signatures is influenced by limited data availability, which may restrict statistical power. Moreover, methods for integration of large sample cohorts and signature identification are limited. We present a step-by-step computational protocol for functional gene expression analysis and the identification of diagnostic and prognostic signatures by combining meta-analysis with machine learning and survival analysis. The novelty of the toolbox lies in its all-in-one functionality, generic design, and modularity. It is exemplified for lung cancer, including a comprehensive evaluation using different validation strategies. However, the protocol is not restricted to specific disease types and can therefore be used by a broad community. The accompanying R package vignette runs in ~1 h and describes the workflow in detail for use by researchers with limited bioinformatics training.
Collapse
|
5
|
Notarnicola M, Tutino V, Tafaro A, Bianco G, Guglielmi E, Caruso MG. Dietary olive oil induces cannabinoid CB2 receptor expression in adipose tissue of Apc Min/+ transgenic mice. ACTA ACUST UNITED AC 2016; 4:73-80. [PMID: 28035344 PMCID: PMC5166557 DOI: 10.3233/nha-160008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND: Cannabinoid- 2 (CB2) receptor is known for its anti-obesity effects silencing the activated immune cells that are key drivers of metabolic syndrome and inflammation. Nutritional interventions in experimental models of carcinogenesis have been demonstrated to modulate tissue inflammation state and proliferation. OBJECTIVE: Aim of this study was to test, in ApcMin/+ mice, whether a diet enriched with olive oil, omega- 3 and omega-6- PUFAs affects the adipose tissue inflammation status. METHODS: Four groups of animal were studied: ST group, receiving a standard diet; OO group, receiving the standard diet in which soybean oil (source of fats) was replaced with olive oil; OM-3 group, receiving the standard diet in which soybean oil was replaced with salmon oil; OM-6 group, receiving the standard diet in which soybean oil was replaced with oenothera oil. Gene and protein expression, in adipose tissue, were evaluated by RT-PCR and Western Blotting, respectively. Enzymatic activities were assayed by fluorescent and radiometric method, where appropriated. RESULTS: The diet enriched with olive oil significantly induced CB2 receptor expression and it was able to control inflammatory and proliferative activity of mice adipose tissue. CONCLUSIONS: The present findings open opportunities for developing novel nutritional strategies considering olive oil a key ingredient of a healthy dietary pattern.
Collapse
Affiliation(s)
- Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| | - Valeria Tutino
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| | - Angela Tafaro
- Laboratory of Animal Facility, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| | - Giusy Bianco
- Laboratory of Animal Facility, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| | - Emilia Guglielmi
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| | - Maria Gabriella Caruso
- Laboratory of Nutritional Biochemistry, National Institute for Digestive Diseases "S. de Bellis" , Castellana Grotte, Bari, Italy
| |
Collapse
|
6
|
Uno K, Yamada T, Ishigaki Y, Imai J, Hasegawa Y, Sawada S, Kaneko K, Ono H, Asano T, Oka Y, Katagiri H. A hepatic amino acid/mTOR/S6K-dependent signalling pathway modulates systemic lipid metabolism via neuronal signals. Nat Commun 2015; 6:7940. [PMID: 26268630 PMCID: PMC4557134 DOI: 10.1038/ncomms8940] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Metabolism is coordinated among tissues and organs via neuronal signals. Levels of circulating amino acids (AAs), which are elevated in obesity, activate the intracellular target of rapamycin complex-1 (mTORC1)/S6kinase (S6K) pathway in the liver. Here we demonstrate that hepatic AA/mTORC1/S6K signalling modulates systemic lipid metabolism via a mechanism involving neuronal inter-tissue communication. Hepatic expression of an AA transporter, SNAT2, activates the mTORC1/S6K pathway, and markedly elevates serum triglycerides (TGs), while downregulating adipose lipoprotein lipase (LPL). Hepatic Rheb or active-S6K expression have similar metabolic effects, whereas hepatic expression of dominant-negative-S6K inhibits TG elevation in SNAT2 mice. Denervation, pharmacological deafferentation and β-blocker administration suppress obesity-related hypertriglyceridemia with adipose LPL upregulation, suggesting that signals are transduced between liver and adipose tissue via a neuronal pathway consisting of afferent vagal and efferent sympathetic nerves. Thus, the neuronal mechanism uncovered here serves to coordinate amino acid and lipid levels and contributes to the development of obesity-related hypertriglyceridemia.
Collapse
Affiliation(s)
- Kenji Uno
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yasushi Ishigaki
- Division of Diabetes and Metabolism, Iwate Medical University, Morioka 020-8505, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yutaka Hasegawa
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hiraku Ono
- The Fourth Department of Internal Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, University of Hiroshima, Hiroshima 734-8553, Japan
| | - Yoshitomo Oka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.,Japan Science and Technology Agency, CREST, Sendai 980-8575, Japan
| |
Collapse
|
7
|
Guardiola-Serrano F, Beteta-Göbel R, Rodríguez-Lorca R, Ibarguren M, López DJ, Terés S, Alvarez R, Alonso-Sande M, Busquets X, Escribá PV. The Novel Anticancer Drug Hydroxytriolein Inhibits Lung Cancer Cell Proliferation via a Protein Kinase Cα- and Extracellular Signal-Regulated Kinase 1/2-Dependent Mechanism. J Pharmacol Exp Ther 2015; 354:213-24. [PMID: 26065701 DOI: 10.1124/jpet.114.222281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/09/2015] [Indexed: 03/08/2025] Open
Abstract
Membrane lipid therapy is a novel approach to rationally design or discover therapeutic molecules that target membrane lipids. This strategy has been used to design synthetic fatty acid analogs that are currently under study in clinical trials for the treatment of cancer. In this context, and with the aim of controlling tumor cell growth, we have designed and synthesized a hydroxylated analog of triolein, hydroxytriolein (HTO). Both triolein and HTO regulate the biophysical properties of model membranes, and they inhibit the growth of non-small-cell lung cancer (NSCLC) cell lines in vitro. The molecular mechanism underlying the antiproliferative effect of HTO involves regulation of the lipid membrane structure, protein kinase C-α and extracellular signal-regulated kinase activation, the production of reactive oxygen species, and autophagy. In vivo studies on a mouse model of NSCLC showed that HTO, but not triolein, impairs tumor growth, which could be associated with the relative resistance of HTO to enzymatic degradation. The data presented explain in part why olive oil (whose main component is the triacylglycerol triolein) is preventive but not therapeutic, and they demonstrate a potent effect of HTO against cancer. HTO shows a good safety profile, it can be administered orally, and it does not induce nontumor cell (fibroblast) death in vitro or side effects in mice, reflecting its specificity for cancer cells. For these reasons, HTO is a good candidate as a drug to combat cancer that acts by regulating lipid structure and function in the cancer cell membrane.
Collapse
Affiliation(s)
- Francisca Guardiola-Serrano
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.).
| | - Roberto Beteta-Göbel
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Raquel Rodríguez-Lorca
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Maitane Ibarguren
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - David J López
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Silvia Terés
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Rafael Alvarez
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - María Alonso-Sande
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Xavier Busquets
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| | - Pablo V Escribá
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain (F.G.-S., R.B.-G., R.R.-L., M.I., D.J.L., S.T., R.A., M.A.-S., X.B., P.V.E.); and Lipopharma Therapeutics S.L., Palma de Mallorca, Spain (M.I., D.J.L., S.T., R.A., M.A.-S.)
| |
Collapse
|
8
|
Obesity and cancer progression: is there a role of fatty acid metabolism? BIOMED RESEARCH INTERNATIONAL 2015; 2015:274585. [PMID: 25866768 PMCID: PMC4383231 DOI: 10.1155/2015/274585] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/24/2014] [Indexed: 12/30/2022]
Abstract
Currently, there is renewed interest in elucidating the metabolic characteristics of cancer and how these characteristics may be exploited as therapeutic targets. Much attention has centered on glucose, glutamine and de novo lipogenesis, yet the metabolism of fatty acids that arise from extracellular, as well as intracellular, stores as triacylglycerol has received much less attention. This review focuses on the key pathways of fatty acid metabolism, including uptake, esterification, lipolysis, and mitochondrial oxidation, and how the regulators of these pathways are altered in cancer. Additionally, we discuss the potential link that fatty acid metabolism may serve between obesity and changes in cancer progression.
Collapse
|
9
|
Kersten S. Physiological regulation of lipoprotein lipase. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:919-33. [PMID: 24721265 DOI: 10.1016/j.bbalip.2014.03.013] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/27/2014] [Accepted: 03/30/2014] [Indexed: 01/01/2023]
Abstract
The enzyme lipoprotein lipase (LPL), originally identified as the clearing factor lipase, hydrolyzes triglycerides present in the triglyceride-rich lipoproteins VLDL and chylomicrons. LPL is primarily expressed in tissues that oxidize or store fatty acids in large quantities such as the heart, skeletal muscle, brown adipose tissue and white adipose tissue. Upon production by the underlying parenchymal cells, LPL is transported and attached to the capillary endothelium by the protein GPIHBP1. Because LPL is rate limiting for plasma triglyceride clearance and tissue uptake of fatty acids, the activity of LPL is carefully controlled to adjust fatty acid uptake to the requirements of the underlying tissue via multiple mechanisms at the transcriptional and post-translational level. Although various stimuli influence LPL gene transcription, it is now evident that most of the physiological variation in LPL activity, such as during fasting and exercise, appears to be driven via post-translational mechanisms by extracellular proteins. These proteins can be divided into two main groups: the liver-derived apolipoproteins APOC1, APOC2, APOC3, APOA5, and APOE, and the angiopoietin-like proteins ANGPTL3, ANGPTL4 and ANGPTL8, which have a broader expression profile. This review will summarize the available literature on the regulation of LPL activity in various tissues, with an emphasis on the response to diverse physiological stimuli.
Collapse
Affiliation(s)
- Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703HD Wageningen, The Netherlands
| |
Collapse
|
10
|
Zinrajh D, Hörl G, Jürgens G, Marc J, Sok M, Cerne D. Increased phosphatidylethanolamine N-methyltransferase gene expression in non-small-cell lung cancer tissue predicts shorter patient survival. Oncol Lett 2014; 7:2175-2179. [PMID: 24932311 PMCID: PMC4049682 DOI: 10.3892/ol.2014.2035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/14/2014] [Indexed: 01/31/2023] Open
Abstract
Lipid mobilization is of great importance for tumor growth and studies have suggested that cancer cells exhibit abnormal choline phospholipid metabolism. In the present study, we hypothesized that phosphatidylethanolamine N-methyltransferase (PEMT) gene expression is increased in non-small-cell lung cancer (NSCLC) tissues and that increased gene expression acts as a predictor of shorter patient survival. Forty-two consecutive patients with resected NSCLC were enrolled in this study. Paired samples of lung cancer tissues and adjacent non-cancer lung tissues were collected from resected specimens for the estimation of PEMT expression. SYBR Green-based real-time polymerase chain reaction was used for quantification of PEMT mRNA in lung cancer tissues. Lipoprotein lipase (LPL) and fatty acid synthase (FASN) activities had already been measured in the same tissues. During a four-year follow-up, 21 patients succumbed to tumor progression. One patient did not survive due to non-cancer reasons and was not included in the analysis. Cox regression analysis was used to assess the prognostic value of PEMT expression. Our findings show that elevated PEMT expression in the cancer tissue, relative to that in the adjacent non-cancer lung tissue, predicts shorter patient survival independently of standard prognostic factors and also independently of increased LPL or FASN activity, the two other lipid-related predictors of shorter patient survival. These findings suggest that active phosphatidylcholine and/or choline metabolism are essential for tumor growth and progression.
Collapse
Affiliation(s)
- David Zinrajh
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Gerd Hörl
- Department of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Günther Jürgens
- Department of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz, A-8010 Graz, Austria
| | - Janja Marc
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Miha Sok
- Department of Thoracic Surgery, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Darko Cerne
- Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Podgornik H, Sok M, Kern I, Marc J, Cerne D. Lipoprotein lipase in non-small cell lung cancer tissue is highly expressed in a subpopulation of tumor-associated macrophages. Pathol Res Pract 2013; 209:516-20. [PMID: 23880163 DOI: 10.1016/j.prp.2013.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/13/2012] [Accepted: 06/18/2013] [Indexed: 01/18/2023]
Abstract
High lipoprotein lipase (LPL) activity in non-small cell lung cancer (NSCLC) tissue strongly predicts shorter patient survival. We tested the hypothesis that in NSCLC tissue, macrophages are the major site of LPL expression. LPL expression in the entire NSCLC tissue and in the adjacent non-cancer lung tissue was compared to the expression of genes preferentially expressed in macrophages. LPL expression at the cellular level was analyzed by mRNA fluorescence in situ hybridization. In the whole cancer tissue (but not in the adjacent non-cancer tissue), expression of LPL correlated with expression of genes preferentially expressed in macrophages (MSR1, CD163, FOLR2), but not with expression of genes preferentially expressed in tumor cells. All cells in the cancer and adjacent non-cancer tissue exhibit low LPL expression. However, in cancer tissue only, there were individual highly LPL-expressing cells which were macrophages. These LPL-overexpressing cells were approximately 10 times less abundant than anti-CD163-stained, tumor-associated macrophages. To conclude, in NSCLC tissue, a subpopulation of tumor-associated macrophages highly expresses LPL. Because tumor-associated macrophages are pro-tumorigenic, these cells should be further characterized to better understand the underlying nature of the close relationship between high LPL activity in NSCLC tissue and shorter patient survival.
Collapse
Affiliation(s)
- Helena Podgornik
- Department of Haematology, University Medical Centre Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
12
|
Notarnicola M, Misciagna G, Tutino V, Chiloiro M, Osella AR, Guerra V, Bonfiglio C, Caruso MG. Increased serum levels of lipogenic enzymes in patients with severe liver steatosis. Lipids Health Dis 2012; 11:145. [PMID: 23110339 PMCID: PMC3494566 DOI: 10.1186/1476-511x-11-145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 10/26/2012] [Indexed: 01/21/2023] Open
Abstract
Background Lipid metabolism is altered in subjects with liver steatosis. FAS is a key enzyme in de novo lipogenesis and both FAS gene expression and enzymatic activity are primarily regulated by metabolic signals in the liver. Lipoprotein lipase (LPL), the rate-limiting enzyme for the hydrolysis of core triglycerides, plays a pivotal role in lipid metabolism. This study aims to investigate if circulating levels of FAS and LPL could be clinically associated with liver steatosis. Methods In this work, we present data obtained from a subsample of 94 subjects with liver steatosis enrolled by NUTRIEPA study, a nutritional trial in subjects with liver steatosis. Serum levels of FAS protein and LPL activity were evaluated by ELISA test and by a fluorescent method, respectively. The diagnosis and the degree of liver steatosis were based on laboratory and ecographic measurements. Statistical methods included Kruskal-Wallis analysis of variance and Wilcoxon signed-rank test, where appropriate. The χ2 test has been performed to analyse categorical variables. Results The subjects with severe steatosis had significantly higher serum levels of FAS protein and LPL activity compared to subjects with mild and moderate liver steatosis. Moreover, a positive trend in serum levels of FAS expression from lower to higher degree of steatosis was also detected. Conclusions We describe a relationship between human liver steatosis and elevated levels of circulating lipogenic enzymes. Increased serum levels of FAS expression and LPL activity could be considered a marker of severe liver steatosis.
Collapse
Affiliation(s)
- Maria Notarnicola
- Laboratory of Biochemistry, National Institute for Digestive Diseases, Castellana Grotte, 70013, Bari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Notarnicola M, Miccolis A, Tutino V, Lorusso D, Caruso MG. Low levels of lipogenic enzymes in peritumoral adipose tissue of colorectal cancer patients. Lipids 2011; 47:59-63. [PMID: 22090062 DOI: 10.1007/s11745-011-3630-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/02/2011] [Indexed: 12/21/2022]
Abstract
Lipoprotein lipase (LPL) is the crucial enzyme for intravascular catabolism of triglyceride-rich lipoproteins. Fatty acid synthase (FAS) is a key anabolic enzyme that catalyzes the terminal steps in the novo biosynthesis of 18:2n-6. The involvement of both LPL and FAS in tumor biology has been widely demonstrated in different studies and to verify whether there are regional differences in the expression of these enzymes in visceral adipose tissue from patients with colorectal cancer might be representative of events which sustain tumor growth. The objective of this study was to evaluate LPL and FAS activity and expression of their genes in adipose tissue adjacent to neoplasia and distant from it from patients operated for colorectal cancer. LPL enzymatic activity was evaluated by a fluorescent method and FAS activity by a radiometer assay. Reverse-transcription and real-time PCR were used to detect mRNA levels of two enzymes. Our findings show a significant reduction in both LPL and FAS gene expression and activity levels in adipose tissue adjacent to tumor lesion compared to those detected in paired tissue distant from neoplasia. These results underline the influence of tumor microenvironment on lipid metabolism in adipose tissue, demonstrating a tumor-induced impairment in the formation and lipid storing capacity of adipose tissue in patients with colorectal cancer.
Collapse
Affiliation(s)
- Maria Notarnicola
- Laboratory of Biochemistry, National Institute for Digestive Diseases "S. de Bellis", Via Turi, 27, 70013, Castellana Grotte, Bari, Italy.
| | | | | | | | | |
Collapse
|
14
|
Cerne D, Zitnik IP, Sok M. Increased fatty acid synthase activity in non-small cell lung cancer tissue is a weaker predictor of shorter patient survival than increased lipoprotein lipase activity. Arch Med Res 2011; 41:405-9. [PMID: 21044743 DOI: 10.1016/j.arcmed.2010.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 07/21/2010] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Cumulative evidence suggests the involvement of fatty acid synthase (FAS) in tumor growth. We tested the hypothesis that increased FAS activity and gene expression in non-small cell lung cancer (NSCLC) tissue have a prognostic significance that is independent of that of increased lipoprotein lipase (LPL) activity in the same tissue. METHODS Forty two consecutive patients with resected NSCLC were enrolled in the study. Paired samples of lung cancer tissue and adjacent non-cancer lung tissue were collected from resected specimens for estimation of FAS activity and expression of its gene. LPL activity had previously been measured in the same tissues. During a 4-year follow-up, 21 patients died due to tumor progression. One patient died due to a non-cancer reason and was not included in the analysis. RESULTS High FAS activity in cancerous tissue relative to that in the adjacent non-cancer lung tissue was associated with weight loss in the 3 months immediately before tumor excision and patient death during the follow-up. Higher FAS activity in the cancer tissue was associated with higher LPL activity in the same tissue, which also predicted shorter patient survival, but LPL was the stronger predictor. FAS gene expression was higher in the adjacent non-cancer tissue than in the cancer tissue but had no predictive value. CONCLUSION Our study further underlines the involvement of cancer tissue FAS activity in tumor growth but also indicates its weaker importance compared to LPL activity.
Collapse
Affiliation(s)
- Darko Cerne
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| | | | | |
Collapse
|
15
|
Staege MS, Hesse M, Max D. Lipases and Related Molecules in Cancer. CANCER GROWTH AND METASTASIS 2010. [DOI: 10.4137/cgm.s2816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lipases are enzymes that catalyze the hydrolysis of lipids. Based on protein structures and sequences, lipases can be classified into different protein families. The majority of conventional mammalian lipases are members of the protein super-families of serine esterases and alpha-beta hydrolases. Differential expression of lipases and related alpha-beta hydrolases in tumor cells has been observed. The physiological or patho-physiological functions of these tumor related enzymes are largely unknown. However, lipases are not only involved in energy metabolism but also in the metabolism of bioactive molecules, e.g. phosphatidic acid or arachidonic acid, suggesting that tumor-specifically expressed lipases might be interesting targets for the development of future treatment strategies. Moreover, independent of the patho-physiological function, tumor associated lipases can serve as targets for immunological treatment strategies. In addition, lipases with exclusive expression in single tumor entities can serve as potential diagnostic targets.
Collapse
Affiliation(s)
- Martin S. Staege
- Martin-Luther-University Halle-Wittenberg, University Clinic and Polyclinic for Child and Adolescent Medicine, D-06097 Halle, Germany
| | - Manuela Hesse
- Martin-Luther-University Halle-Wittenberg, University Clinic and Polyclinic for Child and Adolescent Medicine, D-06097 Halle, Germany
| | - Daniela Max
- Martin-Luther-University Halle-Wittenberg, University Clinic and Polyclinic for Child and Adolescent Medicine, D-06097 Halle, Germany
| |
Collapse
|
16
|
Wiwanitkit V. Lipoprotein Lipase Activity and Prediction for Survival in Lung Cancer. Arch Med Res 2010; 41:64. [DOI: 10.1016/j.arcmed.2009.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 10/12/2009] [Indexed: 11/28/2022]
|