55 results on '"Dehairs J"'
Search Results
2. ApoA-I mimetic peptide 5A boosts remyelination by promoting myelin debris clearance
- Author
-
VANHERLE, Sam, GERVOIS, Pascal, DIERCKX, Tess, LOIX, Melanie, JORISSEN, Winde, Dehairs, J., Swinnen, J., Mulder, M., Remaley, A.T., LAMBRICHTS, Ivo, HENDRIKS, Jerome, HAIDAR, Mansour, BOGIE, Jeroen, VANHERLE, Sam, GERVOIS, Pascal, DIERCKX, Tess, LOIX, Melanie, JORISSEN, Winde, Dehairs, J., Swinnen, J., Mulder, M., Remaley, A.T., LAMBRICHTS, Ivo, HENDRIKS, Jerome, HAIDAR, Mansour, and BOGIE, Jeroen
- Subjects
ApoA-I mimetic peptide 5A ,phagocytere ,myelination ,myelin debris clearance ,lipid droplet degradation - Abstract
Article The ApoA-I mimetic peptide 5A enhances remyelination by promoting clearance and degradation of myelin debris Graphical abstract Highlights d ApoA-I mimetic peptide 5A enhances remyelination in a phagocyte-dependent manner d In addition to promoting lipid efflux, peptide 5A enhances clearance of myelin debris d Peptide 5A drives clearance of myelin debris via the fatty acid translocase CD36 We thank M.P. Tulleners for excellent technical assistance. The work was financially supported by the Research Foundation of Flanders (FWO Vlaanderen; 1S15519N, G099618FWO, and 12J9119N) and the Interreg V-A EMR program (EURLIPIDS, EMR23). The funding agencies had no role in the design, analysis, or writing of the article.
- Published
- 2021
3. Sustained SREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy : SREBP-1-afhankelijke lipogenese als een sleutelmechanisme in resistentie tegen doelgerichte BRAF therapie
- Author
-
Talebi, A, Dehairs, J, Rambow, F, Rogiers, A, Nittner, D, Derua, R, Vanderhoydonc, F, Duarte, JAG, Bosisio, F, Van den Eynde, K, Nys, K, Pérez, MV, Agostinis, P, Waelkens, E, Van den Oord, J, Fendt, SM, Marine, JC, Swinnen, JV, Munck, Sebastian, and Swinnen, Johan
- Subjects
lipids ,melanoma ,SREBP ,BRAF - Abstract
Whereas significant anti-tumor responses are observed in most BRAFV600E-mutant melanoma patients exposed to MAPK-targeting agents, resistance almost invariably develops. Here, we show that in therapy-responsive cells BRAF inhibition induces downregulation of the processing of Sterol Regulator Element Binding (SREBP-1) and thereby lipogenesis. Irrespective of the escape mechanism, therapy-resistant cells invariably restore this process to promote lipid saturation and protect melanoma from ROS-induced damage and lipid peroxidation. Importantly, pharmacological SREBP-1 inhibition sensitizes BRAFV600E-mutant therapy-resistant melanoma to BRAFV600E inhibitors both in vitro and in a pre-clinical PDX in vivo model. Together, these data indicate that targeting SREBP-1-induced lipogenesis may offer a new avenue to overcome acquisition of resistance to BRAF-targeted therapy. This work also provides evidence that targeting vulnerabilities downstream of oncogenic signaling offers new possibilities in overcoming resistance to targeted therapies. ispartof: Nature Communications vol:9 issue:1 ispartof: location:England nrpages: 132 status: published
- Published
- 2018
4. Fat Induces Glucose Metabolism in Nontransformed Liver Cells and Promotes Liver Tumorigenesis
- Author
-
Rebeca Alba Rubio, Shinya Kuroda, Chantal Mathieu, Yasuaki Karasawa, Jos van Pelt, Jia Zeng, Roberta Schmieder, Thomas G. P. Grunewald, Johannes V. Swinnen, Bryan Holvoet, Jonas Dehairs, Masashi Fujii, Roman Vangoitsenhoven, Francesco Napolitano, Diego di Bernardo, James Dooley, Koen Veys, Adrian Liston, Dorien Broekaert, Lindsay A. Broadfield, Juan Fernández-García, Sarah-Maria Fendt, Kim Vriens, Miki Eto, Diether Lambrechts, Joao A.G. Duarte, Katrien De Bock, Suguru Fujita, Christophe Deroose, Mélanie Planque, Joke Van Elsen, Broadfield, L. A., Duarte, J. A. G., Schmieder, R., Broekaert, D., Veys, K., Planque, M., Vriens, K., Karasawa, Y., Napolitano, F., Fujita, S., Fujii, M., Eto, M., Holvoet, B., Vangoitsenhoven, R., Fernandez-Garcia, J., Van Elsen, J., Dehairs, J., Zeng, J., Dooley, J., Rubio, R. A., Van Pelt, J., Grunewald, T. G. P., Liston, A., Mathieu, C., Deroose, C. M., Swinnen, J. V., Lambrechts, D., Di Bernardo, D., Kuroda, S., De Bock, K., and Fendt, S. -M.
- Subjects
0301 basic medicine ,Proteomics ,Cancer Research ,Glucose uptake ,Palmitates ,Palmitate ,Mice ,Random Allocation ,0302 clinical medicine ,Serine ,Hepatocyte ,Dietary Fat ,chemistry.chemical_classification ,Chemistry ,Fatty Acids ,Liver Neoplasms ,3. Good health ,Cell Transformation, Neoplastic ,Oncology ,Liver Neoplasm ,030220 oncology & carcinogenesis ,Pyruvate carboxylase activity ,Liver cancer ,Reactive Oxygen Specie ,Human ,Transcriptional Activation ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Citric Acid Cycle ,Carbohydrate metabolism ,Peroxisome ,Diet, High-Fat ,Article ,03 medical and health sciences ,Internal medicine ,Diabetes mellitus ,medicine ,Peroxisomes ,Animals ,Humans ,Lactic Acid ,Obesity ,Carcinogen ,Pyruvate Carboxylase ,Reactive oxygen species ,Animal ,Proteomic ,Lipid metabolism ,Glucose Tolerance Test ,medicine.disease ,Lipid Metabolism ,Dietary Fats ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Glucose ,Hepatocytes ,Reactive Oxygen Species ,Fatty Acid - Abstract
Hepatic fat accumulation is associated with diabetes and hepatocellular carcinoma (HCC). Here, we characterize the metabolic response that high-fat availability elicits in livers before disease development. After a short term on a high-fat diet (HFD), otherwise healthy mice showed elevated hepatic glucose uptake and increased glucose contribution to serine and pyruvate carboxylase activity compared with control diet (CD) mice. This glucose phenotype occurred independently from transcriptional or proteomic programming, which identifies increased peroxisomal and lipid metabolism pathways. HFD-fed mice exhibited increased lactate production when challenged with glucose. Consistently, administration of an oral glucose bolus to healthy individuals revealed a correlation between waist circumference and lactate secretion in a human cohort. In vitro, palmitate exposure stimulated production of reactive oxygen species and subsequent glucose uptake and lactate secretion in hepatocytes and liver cancer cells. Furthermore, HFD enhanced the formation of HCC compared with CD in mice exposed to a hepatic carcinogen. Regardless of the dietary background, all murine tumors showed similar alterations in glucose metabolism to those identified in fat exposed nontransformed mouse livers, however, particular lipid species were elevated in HFD tumor and nontumor-bearing HFD liver tissue. These findings suggest that fat can induce glucose-mediated metabolic changes in nontransformed liver cells similar to those found in HCC. Significance: With obesity-induced hepatocellular carcinoma on a rising trend, this study shows in normal, nontransformed livers that fat induces glucose metabolism similar to an oncogenic transformation.
- Published
- 2021
5. Intrinsic temperature increase drives lipid metabolism towards ferroptosis evasion and chemotherapy resistance in pancreatic cancer.
- Author
-
de Laat V, Topal H, Spotbeen X, Talebi A, Dehairs J, Idkowiak J, Vanderhoydonc F, Ostyn T, Zhao P, Jacquemyn M, Wölk M, Sablina A, Augustyns K, Vanden Berghe T, Roskams T, Daelemans D, Fedorova M, Topal B, and Swinnen JV
- Subjects
- Humans, Cell Line, Tumor, Lipid Peroxidation drug effects, p38 Mitogen-Activated Protein Kinases metabolism, Temperature, Animals, Mice, Ferroptosis drug effects, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Drug Resistance, Neoplasm genetics, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal genetics, Gemcitabine, Lipid Metabolism drug effects, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use
- Abstract
A spontaneously occurring temperature increase in solid tumors has been reported sporadically, but is largely overlooked in terms of cancer biology. Here we show that temperature is increased in tumors of patients with pancreatic ductal adenocarcinoma (PDAC) and explore how this could affect therapy response. By mimicking this observation in PDAC cell lines, we demonstrate that through adaptive changes in lipid metabolism, the temperature increase found in human PDAC confers protection to lipid peroxidation and contributes to gemcitabine resistance. Consistent with the recently uncovered role of p38 MAPK in ferroptotic cell death, we find that the reduction in lipid peroxidation potential following adaptation to tumoral temperature allows for p38 MAPK inhibition, conferring chemoresistance. As an increase in tumoral temperature is observed in several other tumor types, our findings warrant taking tumoral temperature into account in subsequent studies related to ferroptosis and therapy resistance. More broadly, our findings indicate that tumoral temperature affects cancer biology., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. The Effect of FTY720 on Sphingolipid Imbalance and Cognitive Decline in Aged EFAD Mice.
- Author
-
Luo Q, Crivelli SM, Zong S, Giovagnoni C, van Kruining D, Mané-Damas M, den Hoedt S, Berkes D, De Vries HE, Mulder MT, Walter J, Waelkens E, Derua R, Swinnen JV, Dehairs J, Losen M, and Martinez-Martinez P
- Abstract
Background: During Alzheimer's disease (AD) progression, there is a decline in the bioactive sphingolipid sphingosine-1-phosphate (S1P). Previous research showed that FTY720, an S1P mimetic, prevented cognitive decline and reduced ceramide levels in transgenic mice with familial AD carrying the human APOE4 gene (E4FAD) at 6-7 months of age., Objective: The objective of this study is to explore the protective effects of FTY720 at late-stage AD., Methods: Male mice aged 9.5 to 10.5 months were orally administered FTY720 (0.1 mg/kg) via oral gavage for 6 weeks. A pre-test of water maze was used for evaluating the pathological status. After 4 weeks of administration, memory, locomotion, and anxiety were assessed. Cortex samples were analyzed for amyloid-β (Aβ) and sphingolipid levels., Results: Compared with APOE3 mice, APOE4, E3FAD and E4FAD mice exhibited significant memory deficits. After 6 weeks administration, FTY720 did not alleviate memory deficits in EFAD mice. Lipid analysis revealed that S1P was significantly reduced in EFAD mice (E3FAD or E4FAD) compared to controls (APOE3 and APOE4). Ceramide level alterations were predominantly dependent on APOE isoforms rather than AD transgenes. Interestingly, Cer (d18 : 1/22 : 1) was elevated in APOE4 mice compared to APOE3, and FTY720 reduced it., Conclusions: E4FAD and APOE4 mice exhibited significant spatial memory deficits and higher ceramide concentrations compared to APOE3 mice. FTY720 did not reverse memory deficits in E4FAD and APOE4 mice but reduced specific ceramide species. This study provides insights into the association between sphingolipids and APOE4 in advanced AD stages, exploring potential therapeutic targeting of sphingolipid metabolism., Competing Interests: The authors have no conflict of interest to report., (© 2024 – The authors. Published by IOS Press.)
- Published
- 2024
- Full Text
- View/download PDF
7. PMP22 duplication dysregulates lipid homeostasis and plasma membrane organization in developing human Schwann cells.
- Author
-
Prior R, Silva A, Vangansewinkel T, Idkowiak J, Tharkeshwar AK, Hellings TP, Michailidou I, Vreijling J, Loos M, Koopmans B, Vlek N, Agaser C, Kuipers TB, Michiels C, Rossaert E, Verschoren S, Vermeire W, de Laat V, Dehairs J, Eggermont K, van den Biggelaar D, Bademosi AT, Meunier FA, vandeVen M, Van Damme P, Mei H, Swinnen JV, Lambrichts I, Baas F, Fluiter K, Wolfs E, and Van Den Bosch L
- Subjects
- Animals, Humans, Mice, Gene Duplication, Sciatic Nerve metabolism, Cell Membrane metabolism, Charcot-Marie-Tooth Disease genetics, Charcot-Marie-Tooth Disease metabolism, Charcot-Marie-Tooth Disease pathology, Homeostasis physiology, Induced Pluripotent Stem Cells metabolism, Lipid Metabolism physiology, Myelin Proteins metabolism, Myelin Proteins genetics, Schwann Cells metabolism
- Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is the most common inherited peripheral neuropathy caused by a 1.5 Mb tandem duplication of chromosome 17 harbouring the PMP22 gene. This dose-dependent overexpression of PMP22 results in disrupted Schwann cell myelination of peripheral nerves. To obtain better insights into the underlying pathogenic mechanisms in CMT1A, we investigated the role of PMP22 duplication in cellular homeostasis in CMT1A mouse models and in patient-derived induced pluripotent stem cells differentiated into Schwann cell precursors (iPSC-SCPs). We performed lipidomic profiling and bulk RNA sequencing (RNA-seq) on sciatic nerves of two developing CMT1A mouse models and on CMT1A patient-derived iPSC-SCPs. For the sciatic nerves of the CMT1A mice, cholesterol and lipid metabolism was downregulated in a dose-dependent manner throughout development. For the CMT1A iPSC-SCPs, transcriptional analysis unveiled a strong suppression of genes related to autophagy and lipid metabolism. Gene ontology enrichment analysis identified disturbances in pathways related to plasma membrane components and cell receptor signalling. Lipidomic analysis confirmed the severe dysregulation in plasma membrane lipids, particularly sphingolipids, in CMT1A iPSC-SCPs. Furthermore, we identified reduced lipid raft dynamics, disturbed plasma membrane fluidity and impaired cholesterol incorporation and storage, all of which could result from altered lipid storage homeostasis in the patient-derived CMT1A iPSC-SCPs. Importantly, this phenotype could be rescued by stimulating autophagy and lipolysis. We conclude that PMP22 duplication disturbs intracellular lipid storage and leads to a more disordered plasma membrane owing to an alteration in the lipid composition, which might ultimately lead to impaired axo-glial interactions. Moreover, targeting lipid handling and metabolism could hold promise for the treatment of patients with CMT1A., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2024
- Full Text
- View/download PDF
8. ACSM1 and ACSM3 Regulate Fatty Acid Metabolism to Support Prostate Cancer Growth and Constrain Ferroptosis.
- Author
-
Shrestha RK, Nassar ZD, Hanson AR, Iggo R, Townley SL, Dehairs J, Mah CY, Helm M, Alizadeh-Ghodsi M, Pickering M, Ghesquière B, Watt MJ, Quek LE, Hoy AJ, Tilley WD, Swinnen JV, Butler LM, and Selth LA
- Subjects
- Male, Humans, Animals, Mice, Cell Line, Tumor, Receptors, Androgen metabolism, Lipid Metabolism, Xenograft Model Antitumor Assays, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Prostatic Neoplasms genetics, Coenzyme A Ligases metabolism, Coenzyme A Ligases genetics, Ferroptosis, Fatty Acids metabolism, Cell Proliferation
- Abstract
Solid tumors are highly reliant on lipids for energy, growth, and survival. In prostate cancer, the activity of the androgen receptor (AR) is associated with reprogramming of lipid metabolic processes. Here, we identified acyl-CoA synthetase medium chain family members 1 and 3 (ACSM1 and ACSM3) as AR-regulated mediators of prostate cancer metabolism and growth. ACSM1 and ACSM3 were upregulated in prostate tumors compared with nonmalignant tissues and other cancer types. Both enzymes enhanced proliferation and protected prostate cancer cells from death in vitro, whereas silencing ACSM3 led to reduced tumor growth in an orthotopic xenograft model. ACSM1 and ACSM3 were major regulators of the prostate cancer lipidome and enhanced energy production via fatty acid oxidation. Metabolic dysregulation caused by loss of ACSM1/3 led to mitochondrial oxidative stress, lipid peroxidation, and cell death by ferroptosis. Conversely, elevated ACSM1/3 activity enabled prostate cancer cells to survive toxic levels of medium chain fatty acids and promoted resistance to ferroptosis-inducing drugs and AR antagonists. Collectively, this study reveals a tumor-promoting function of medium chain acyl-CoA synthetases and positions ACSM1 and ACSM3 as key players in prostate cancer progression and therapy resistance. Significance: Androgen receptor-induced ACSM1 and ACSM3 mediate a metabolic pathway in prostate cancer that enables the utilization of medium chain fatty acids for energy production, blocks ferroptosis, and drives resistance to clinically approved antiandrogens., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
9. Circulating Lipid Profiles Associated With Resistance to Androgen Deprivation Therapy in Localized Prostate Cancer.
- Author
-
Lin HM, Yang X, Centenera MM, Huynh K, Giles C, Dehairs J, Swinnen JV, Hoy AJ, Meikle PJ, Butler LM, Taplin ME, and Horvath LG
- Subjects
- Humans, Male, Drug Resistance, Neoplasm, Aged, Lipids blood, Middle Aged, Lipidomics, Nitriles therapeutic use, Prostatectomy, Androgen Antagonists therapeutic use, Prostatic Neoplasms drug therapy, Prostatic Neoplasms blood, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery
- Abstract
Purpose: Intense androgen deprivation therapy (ADT) with androgen receptor pathway inhibitors (ARPIs) before radical prostatectomy (RP) produced favorable pathologic responses in approximately 20% of patients. The molecular reason for the low rate of response remains unclear. Lipid metabolism is known to influence androgen receptor signaling and ARPI efficacy. The aim of the study was to identify circulating lipid profiles associated with ADT/ARPI resistance in localized prostate cancer., Materials and Methods: Two independent experimental approaches were used. Experiment 1: Post hoc analysis of the association between plasma lipidomic profiles and ADT/ARPI response was performed on patients (n = 104) from two phase II trials of neoadjuvant ADT/ARPI. Response to ADT/ARPI was defined by pathologic response. Experiment 2: Patient-derived tumor explants from RP (n = 105) were cultured in enzalutamide for 48 hours. Explant response to enzalutamide was evaluated against pre-RP plasma lipidomic profiles (n = 105) and prostate tissue lipidomic profiles (n = 36). Response was defined by Ki67 (cell proliferation marker) fold difference between enzalutamide and vehicle-treated explants. In both experiments, associations between lipid profiles and ADT/ARPI response were analyzed by latent class analysis., Results: Pretreatment plasma lipid profiles classified each experimental cohort into two groups with differences in ADT/ARPI response rates. The response rates of the groups were 9.6% versus 29% in experiment 1 (chi-squared test P = .012) and 49% versus 70% in experiment 2 (chi-squared test P = .037). In both experiments, the group with a higher incidence of ADT/ARPI resistance had higher plasma levels of sphingomyelin, glycosylceramides, free fatty acids, acylcarnitines, cholesterol esters, and alkyl/alkenyl-phosphatidylcholine and lower plasma levels of triacylglycerols, diacylglycerols, and phosphoethanolamine (t-test P < .05)., Conclusion: Pretreatment circulating lipid profiles are associated with ADT/ARPI resistance in localized cancer in both human cohorts and explant models.
- Published
- 2024
- Full Text
- View/download PDF
10. The gluconeogenesis enzyme PCK2 has a non-enzymatic role in proteostasis in endothelial cells.
- Author
-
de Zeeuw P, Treps L, García-Caballero M, Harjes U, Kalucka J, De Legher C, Brepoels K, Peeters K, Vinckier S, Souffreau J, Bouché A, Taverna F, Dehairs J, Talebi A, Ghesquière B, Swinnen J, Schoonjans L, Eelen G, Dewerchin M, and Carmeliet P
- Subjects
- Humans, Human Umbilical Vein Endothelial Cells metabolism, Glucose metabolism, Autophagy, Unfolded Protein Response, Phosphoenolpyruvate Carboxykinase (ATP), Proteostasis, Gluconeogenesis genetics, Endothelial Cells metabolism, Phosphoenolpyruvate Carboxykinase (GTP) metabolism, Phosphoenolpyruvate Carboxykinase (GTP) genetics
- Abstract
Endothelial cells (ECs) are highly glycolytic, but whether they generate glycolytic intermediates via gluconeogenesis (GNG) in glucose-deprived conditions remains unknown. Here, we report that glucose-deprived ECs upregulate the GNG enzyme PCK2 and rely on a PCK2-dependent truncated GNG, whereby lactate and glutamine are used for the synthesis of lower glycolytic intermediates that enter the serine and glycerophospholipid biosynthesis pathways, which can play key roles in redox homeostasis and phospholipid synthesis, respectively. Unexpectedly, however, even in normal glucose conditions, and independent of its enzymatic activity, PCK2 silencing perturbs proteostasis, beyond its traditional GNG role. Indeed, PCK2-silenced ECs have an impaired unfolded protein response, leading to accumulation of misfolded proteins, which due to defective proteasomes and impaired autophagy, results in the accumulation of protein aggregates in lysosomes and EC demise. Ultimately, loss of PCK2 in ECs impaired vessel sprouting. This study identifies a role for PCK2 in proteostasis beyond GNG., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Peroxisomal β-oxidation enzyme, DECR2, regulates lipid metabolism and promotes treatment resistance in advanced prostate cancer.
- Author
-
Mah CY, Nguyen ADT, Niijima T, Helm M, Dehairs J, Ryan FJ, Ryan N, Quek LE, Hoy AJ, Don AS, Mills IG, Swinnen JV, Lynn DJ, Nassar ZD, and Butler LM
- Subjects
- Male, Humans, Lipid Metabolism genetics, Cell Line, Tumor, Receptors, Androgen genetics, Receptors, Androgen metabolism, Androgens metabolism, Cell Proliferation, Fatty Acids, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant metabolism
- Abstract
Background: Peroxisomes are central metabolic organelles that have key roles in fatty acid homoeostasis. As prostate cancer (PCa) is particularly reliant on fatty acid metabolism, we explored the contribution of peroxisomal β-oxidation (perFAO) to PCa viability and therapy response., Methods: Bioinformatic analysis was performed on clinical transcriptomic datasets to identify the perFAO enzyme, 2,4-dienoyl CoA reductase 2 (DECR2) as a target gene of interest. Impact of DECR2 and perFAO inhibition via thioridazine was examined in vitro, in vivo, and in clinical prostate tumours cultured ex vivo. Transcriptomic and lipidomic profiling was used to determine the functional consequences of DECR2 inhibition in PCa., Results: DECR2 is upregulated in clinical PCa, most notably in metastatic castrate-resistant PCa (CRPC). Depletion of DECR2 significantly suppressed proliferation, migration, and 3D growth of a range of CRPC and therapy-resistant PCa cell lines, and inhibited LNCaP tumour growth and proliferation in vivo. DECR2 influences cell cycle progression and lipid metabolism to support tumour cell proliferation. Further, co-targeting of perFAO and standard-of-care androgen receptor inhibition enhanced suppression of PCa cell proliferation., Conclusion: Our findings support a focus on perFAO, specifically DECR2, as a promising therapeutic target for CRPC and as a novel strategy to overcome lethal treatment resistance., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. 2,8-Dihydroxyadenine-induced nephropathy causes hexosylceramide accumulation with increased mTOR signaling, reduced levels of protective SirT3 expression and impaired renal mitochondrial function.
- Author
-
Moellmann J, Krueger K, Wong DWL, Klinkhammer BM, Buhl EM, Dehairs J, Swinnen JV, Noels H, Jankowski J, Lebherz C, Boor P, Marx N, and Lehrke M
- Subjects
- Animals, Mice, Adenine, Fibrosis, Inflammation metabolism, Kidney pathology, Mice, Inbred C57BL, Mitochondria metabolism, TOR Serine-Threonine Kinases metabolism, Renal Insufficiency, Chronic pathology, Sirtuin 3 genetics, Sirtuin 3 metabolism
- Abstract
Aim: Chronic kidney disease (CKD) is accompanied by increased cardiovascular risk and heart failure (HF). In rodents, 2,8-dihydroxyadenine (DHA)-induced nephropathy is a frequently used CKD model. Cardiac and kidney tubular cells share high energy demand to guarantee constant contractive force of the heart or reabsorption/secretion of primary filtrated molecules and waste products by the kidney. Here we analyze time-dependent mechanisms of kidney damage and cardiac consequences under consideration of energetic pathways with the focus on mitochondrial function and lipid metabolism in mice., Methods and Results: CKD was induced by alternating dietary adenine supplementation (0.2 % or 0.05 % of adenine) in C57BL/6J mice for 9 weeks. Progressive kidney damage led to reduced creatinine clearance, kidney fibrosis and renal inflammation after 3, 6, and 9 weeks. No difference in cardiac function, mitochondrial respiration nor left ventricular fibrosis was observed at any time point. Investigating mechanisms of renal damage, protective SirT3 was decreased in CKD, which contrasted an increase in protein kinase B (AKT) expression, mechanistic target of rapamycin (mTOR) downstream signaling, induction of oxidative and endoplasmic reticulum (ER) stress. This occurred together with impaired renal mitochondrial function and accumulation of hexosylceramides (HexCer) as an established mediator of inflammation and mitochondrial dysfunction in the kidney., Conclusions: 2,8-DHA-induced CKD results in renal activation of the mTOR downstream signaling, endoplasmic reticulum stress, tubular injury, fibrosis, inflammation, oxidative stress and impaired kidney mitochondrial function in conjunction with renal hexosylceramide accumulation in C57BL/6J mice., Competing Interests: Declaration of competing interest NM has given lectures for Boehringer Ingelheim, Sanofi-Aventis, MSD, BMS, AstraZeneca, Lilly, NovoNordisk; has received unrestricted research grants from Boehringer Ingelheim, and has served as an advisor for Bayer, Boehringer Ingelheim, Sanofi-Aventis, MSD, BMS, AstraZeneca, NovoNordisk. In addition, served in trial leadership for Boehringer Ingelheim and NovoNordisk. ML received grants and personal fees from Boehringer Ingelheim, MSD, Novo Nordisk and personal fees from Amgen, Sanofi, Astra Zeneca, Bayer, Lilly, Daiichi Sankyo, Novarits. NM, HN and JJ are founding shareholders of AMICARE Development GmbH. JM, KK, BMK, DWLW, EMB, JD, JVS, CL and PB have nothing to disclose., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Extracellular vesicle-associated cholesterol supports the regenerative functions of macrophages in the brain.
- Author
-
Vanherle S, Guns J, Loix M, Mingneau F, Dierckx T, Wouters F, Kuipers K, Vangansewinkel T, Wolfs E, Lins PP, Bronckaers A, Lambrichts I, Dehairs J, Swinnen JV, Verberk SGS, Haidar M, Hendriks JJA, and Bogie JFJ
- Subjects
- Brain, Macrophages, Cell Differentiation, Cholesterol, Extracellular Vesicles
- Abstract
Macrophages play major roles in the pathophysiology of various neurological disorders, being involved in seemingly opposing processes such as lesion progression and resolution. Yet, the molecular mechanisms that drive their harmful and benign effector functions remain poorly understood. Here, we demonstrate that extracellular vesicles (EVs) secreted by repair-associated macrophages (RAMs) enhance remyelination ex vivo and in vivo by promoting the differentiation of oligodendrocyte precursor cells (OPCs). Guided by lipidomic analysis and applying cholesterol depletion and enrichment strategies, we find that EVs released by RAMs show markedly elevated cholesterol levels and that cholesterol abundance controls their reparative impact on OPC maturation and remyelination. Mechanistically, EV-associated cholesterol was found to promote OPC differentiation predominantly through direct membrane fusion. Collectively, our findings highlight that EVs are essential for cholesterol trafficking in the brain and that changes in cholesterol abundance support the reparative impact of EVs released by macrophages in the brain, potentially having broad implications for therapeutic strategies aimed at promoting repair in neurodegenerative disorders., (© 2023 The Authors. Journal of Extracellular Vesicles published by Wiley Periodicals LLC on behalf of International Society for Extracellular Vesicles.)
- Published
- 2023
- Full Text
- View/download PDF
14. Peroxisome disruption alters lipid metabolism and potentiates antitumor response with MAPK-targeted therapy in melanoma.
- Author
-
Huang F, Cai F, Dahabieh MS, Gunawardena K, Talebi A, Dehairs J, El-Turk F, Park JY, Li M, Goncalves C, Gagnon N, Su J, LaPierre JH, Gaub P, Joyal JS, Mitchell JJ, Swinnen JV, Miller WH Jr, and Del Rincón SV
- Subjects
- Humans, Lipid Metabolism, Ceramides pharmacology, Ceramides metabolism, Peroxisomes metabolism, Melanoma genetics
- Abstract
Melanomas reprogram their metabolism to rapidly adapt to therapy-induced stress conditions, allowing them to persist and ultimately develop resistance. We report that a subpopulation of melanoma cells tolerate MAPK pathway inhibitors (MAPKis) through a concerted metabolic reprogramming mediated by peroxisomes and UDP-glucose ceramide glycosyltransferase (UGCG). Compromising peroxisome biogenesis, by repressing PEX3 expression, potentiated the proapoptotic effects of MAPKis via an induction of ceramides, an effect limited by UGCG-mediated ceramide metabolism. Cotargeting PEX3 and UGCG selectively eliminated a subset of metabolically active, drug-tolerant CD36+ melanoma persister cells, thereby sensitizing melanoma to MAPKis and delaying resistance. Increased levels of peroxisomal genes and UGCG were found in patient-derived MAPKi-relapsed melanomas, and simultaneously inhibiting PEX3 and UGCG restored MAPKi sensitivity in multiple models of therapy resistance. Finally, combination therapy consisting of a newly identified inhibitor of the PEX3-PEX19 interaction, a UGCG inhibitor, and MAPKis demonstrated potent antitumor activity in preclinical melanoma models, thus representing a promising approach for melanoma treatment.
- Published
- 2023
- Full Text
- View/download PDF
15. Sex differences in obesity-induced renal lipid accumulation revealed by lipidomics: a role of adiponectin/AMPK axis.
- Author
-
Juszczak F, Pierre L, Decarnoncle M, Jadot I, Martin B, Botton O, Caron N, Dehairs J, Swinnen JV, and Declèves AE
- Subjects
- Animals, Female, Male, Mice, AMP-Activated Protein Kinases metabolism, Kidney metabolism, Lipidomics, Lipids, Obesity metabolism, Sex Characteristics, Adiponectin, Kidney Diseases etiology
- Abstract
Background: Sex differences have been observed in the development of obesity-related complications in patients, as well as in animal models. Accumulating evidence suggests that sex-dependent regulation of lipid metabolism contributes to sex-specific physiopathology. Lipid accumulation in the renal tissue has been shown to play a major role in the pathogenesis of obesity-induced kidney injury. Unlike in males, the physiopathology of the disease has been poorly described in females, particularly regarding the lipid metabolism adaptation., Methods: Here, we compared the lipid profile changes in the kidneys of female and male mice fed a high-fat diet (HFD) or low-fat diet (LFD) by lipidomics and correlated them with pathophysiological changes., Results: We showed that HFD-fed female mice were protected from insulin resistance and hepatic steatosis compared to males, despite similar body weight gains. Females were particularly protected from renal dysfunction, oxidative stress, and tubular lipid accumulation. Both HFD-fed male and female mice presented dyslipidemia, but lipidomic analysis highlighted differential renal lipid profiles. While both sexes presented similar neutral lipid accumulation with obesity, only males showed increased levels of ceramides and phospholipids. Remarkably, protection against renal lipotoxicity in females was associated with enhanced renal adiponectin and AMP-activated protein kinase (AMPK) signaling. Circulating adiponectin and its renal receptor levels were significantly lower in obese males, but were maintained in females. This observation correlated with the maintained basal AMPK activity in obese female mice compared to males., Conclusions: Collectively, our findings suggest that female mice are protected from obesity-induced renal dysfunction and lipotoxicity associated with enhanced adiponectin and AMPK signaling compared to males., (© 2023. Society for Women's Health Research and BioMed Central Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
16. Fatty acid elongation by ELOVL6 hampers remyelination by promoting inflammatory foam cell formation during demyelination.
- Author
-
Garcia Corrales AV, Verberk SGS, Haidar M, Grajchen E, Dehairs J, Vanherle S, Loix M, Weytjens T, Gervois P, Matsuzaka T, Lambrichts I, Swinnen JV, Bogie JFJ, and Hendriks JJA
- Subjects
- Animals, Mice, Adipogenesis, Disease Models, Animal, Fatty Acids, Fatty Acids, Monounsaturated, Foam Cells, Multiple Sclerosis, Remyelination
- Abstract
A hallmark of multiple sclerosis (MS) is the formation of multiple focal demyelinating lesions within the central nervous system (CNS). These lesions mainly consist of phagocytes that play a key role in lesion progression and remyelination, and therefore represent a promising therapeutic target in MS. We recently showed that unsaturated fatty acids produced by stearoyl-CoA desaturase-1 induce inflammatory foam cell formation during demyelination. These fatty acids are elongated by the "elongation of very long chain fatty acids" proteins (ELOVLs), generating a series of functionally distinct lipids. Here, we show that the expression and activity of ELOVLs are altered in myelin-induced foam cells. Especially ELOVL6, an enzyme responsible for converting saturated and monounsaturated C16 fatty acids into C18 species, was found to be up-regulated in myelin phagocytosing phagocytes in vitro and in MS lesions. Depletion of Elovl6 induced a repair-promoting phagocyte phenotype through activation of the S1P/PPARγ pathway. Elovl6 -deficient foamy macrophages showed enhanced ABCA1-mediated lipid efflux, increased production of neurotrophic factors, and reduced expression of inflammatory mediators. Moreover, our data show that ELOVL6 hampers CNS repair, as Elovl6 deficiency prevented demyelination and boosted remyelination in organotypic brain slice cultures and the mouse cuprizone model. These findings indicate that targeting ELOVL6 activity may be an effective strategy to stimulate CNS repair in MS and other neurodegenerative diseases.
- Published
- 2023
- Full Text
- View/download PDF
17. C/EBPα Confers Dependence to Fatty Acid Anabolic Pathways and Vulnerability to Lipid Oxidative Stress-Induced Ferroptosis in FLT3-Mutant Leukemia.
- Author
-
Sabatier M, Birsen R, Lauture L, Mouche S, Angelino P, Dehairs J, Goupille L, Boussaid I, Heiblig M, Boet E, Sahal A, Saland E, Santos JC, Armengol M, Fernández-Serrano M, Farge T, Cognet G, Simonetta F, Pignon C, Graffeuil A, Mazzotti C, Avet-Loiseau H, Delos O, Bertrand-Michel J, Chedru A, Dembitz V, Gallipoli P, Anstee NS, Loo S, Wei AH, Carroll M, Goubard A, Castellano R, Collette Y, Vergez F, Mansat-De Mas V, Bertoli S, Tavitian S, Picard M, Récher C, Bourges-Abella N, Granat F, Kosmider O, Sujobert P, Colsch B, Joffre C, Stuani L, Swinnen JV, Guillou H, Roué G, Hakim N, Dejean AS, Tsantoulis P, Larrue C, Bouscary D, Tamburini J, and Sarry JE
- Subjects
- Humans, CCAAT-Enhancer-Binding Protein-alpha genetics, CCAAT-Enhancer-Binding Protein-alpha metabolism, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism, Fatty Acids, Mutation, Oxidative Stress, Protein Kinase Inhibitors therapeutic use, Cell Line, Tumor, Ferroptosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
Although transcription factor CCAAT-enhancer binding protein α (C/EBPα) is critical for normal and leukemic differentiation, its role in cell and metabolic homeostasis is largely unknown in cancer. Here, multiomics analyses uncovered a coordinated activation of C/EBPα and Fms-like tyrosine kinase 3 (FLT3) that increased lipid anabolism in vivo and in patients with FLT3-mutant acute myeloid leukemia (AML). Mechanistically, C/EBPα regulated the fatty acid synthase (FASN)-stearoyl-CoA desaturase (SCD) axis to promote fatty acid (FA) biosynthesis and desaturation. We further demonstrated that FLT3 or C/EBPα inactivation decreased monounsaturated FA incorporation to membrane phospholipids through SCD downregulation. Consequently, SCD inhibition enhanced susceptibility to lipid redox stress that was exploited by combining FLT3 and glutathione peroxidase 4 inhibition to trigger lipid oxidative stress, enhancing ferroptotic death of FLT3-mutant AML cells. Altogether, our study reveals a C/EBPα function in lipid homeostasis and adaptation to redox stress, and a previously unreported vulnerability of FLT3-mutant AML to ferroptosis with promising therapeutic application., Significance: FLT3 mutations are found in 30% of AML cases and are actionable by tyrosine kinase inhibitors. Here, we discovered that C/EBPα regulates FA biosynthesis and protection from lipid redox stress downstream mutant-FLT3 signaling, which confers a vulnerability to ferroptosis upon FLT3 inhibition with therapeutic potential in AML. This article is highlighted in the In This Issue feature, p. 1501., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
18. Fatty acid desaturation by stearoyl-CoA desaturase-1 controls regulatory T cell differentiation and autoimmunity.
- Author
-
Grajchen E, Loix M, Baeten P, Côrte-Real BF, Hamad I, Vanherle S, Haidar M, Dehairs J, Broos JY, Ntambi JM, Zimmermann R, Breinbauer R, Stinissen P, Hellings N, Verberk SGS, Kooij G, Giera M, Swinnen JV, Broux B, Kleinewietfeld M, Hendriks JJA, and Bogie JFJ
- Subjects
- Animals, Autoimmunity, Fatty Acids metabolism, Cell Differentiation, Stearoyl-CoA Desaturase genetics, Stearoyl-CoA Desaturase metabolism, Autoimmune Diseases
- Abstract
The imbalance between pathogenic and protective T cell subsets is a cardinal feature of autoimmune disorders such as multiple sclerosis (MS). Emerging evidence indicates that endogenous and dietary-induced changes in fatty acid metabolism have a major impact on both T cell fate and autoimmunity. To date, however, the molecular mechanisms that underlie the impact of fatty acid metabolism on T cell physiology and autoimmunity remain poorly understood. Here, we report that stearoyl-CoA desaturase-1 (SCD1), an enzyme essential for the desaturation of fatty acids and highly regulated by dietary factors, acts as an endogenous brake on regulatory T-cell (Treg) differentiation and augments autoimmunity in an animal model of MS in a T cell-dependent manner. Guided by RNA sequencing and lipidomics analysis, we found that the absence of Scd1 in T cells promotes the hydrolysis of triglycerides and phosphatidylcholine through adipose triglyceride lipase (ATGL). ATGL-dependent release of docosahexaenoic acid enhanced Treg differentiation by activating the nuclear receptor peroxisome proliferator-activated receptor gamma. Our findings identify fatty acid desaturation by SCD1 as an essential determinant of Treg differentiation and autoimmunity, with potentially broad implications for the development of novel therapeutic strategies and dietary interventions for autoimmune disorders such as MS., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. Phospholipase D3 degrades mitochondrial DNA to regulate nucleotide signaling and APP metabolism.
- Author
-
Van Acker ZP, Perdok A, Hellemans R, North K, Vorsters I, Cappel C, Dehairs J, Swinnen JV, Sannerud R, Bretou M, Damme M, and Annaert W
- Subjects
- Mitochondria, Nucleotidyltransferases, Amyloidogenic Proteins, Chromogranin A, Phospholipases, DNA, Mitochondrial, Nucleotides
- Abstract
Phospholipase D3 (PLD3) polymorphisms are linked to late-onset Alzheimer's disease (LOAD). Being a lysosomal 5'-3' exonuclease, its neuronal substrates remained unknown as well as how a defective lysosomal nucleotide catabolism connects to AD-proteinopathy. We identified mitochondrial DNA (mtDNA) as a major physiological substrate and show its manifest build-up in lysosomes of PLD3-defective cells. mtDNA accretion creates a degradative (proteolytic) bottleneck that presents at the ultrastructural level as a marked abundance of multilamellar bodies, often containing mitochondrial remnants, which correlates with increased PINK1-dependent mitophagy. Lysosomal leakage of mtDNA to the cytosol activates cGAS-STING signaling that upregulates autophagy and induces amyloid precursor C-terminal fragment (APP-CTF) and cholesterol accumulation. STING inhibition largely normalizes APP-CTF levels, whereas an APP knockout in PLD3-deficient backgrounds lowers STING activation and normalizes cholesterol biosynthesis. Collectively, we demonstrate molecular cross-talks through feedforward loops between lysosomal nucleotide turnover, cGAS-STING and APP metabolism that, when dysregulated, result in neuronal endolysosomal demise as observed in LOAD., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. Altered Functionality of Lipoprotein(a) Impacts on Angiogenesis in Diabetic Retinopathy.
- Author
-
Shariatzadeh M, Nagtzaam NMA, van Vark-van der Zee L, van Holten-Neelen C, Verhoeven AJM, Dehairs J, Swinnen JV, Leijten F, Ten Berge JC, Ciriano JPM, Wong KT, Mulder M, Leenen PJM, and Dik WA
- Subjects
- Humans, Lipoprotein(a), Intercellular Adhesion Molecule-1, Endothelial Cells, Leukocytes, Mononuclear, Tumor Necrosis Factor-alpha, Vascular Cell Adhesion Molecule-1, Diabetic Retinopathy, Diabetes Mellitus, Type 2 complications
- Abstract
Purpose: Diabetic retinopathy (DR) is a complication of type 2 diabetes mellitus (T2DM). Lipoprotein(a) (Lp(a)) contributes to the progression of DR, but how is unclear. In homeostasis of the retinal microvasculature, myeloid-derived pro-angiogenic cells (PACs) also play a pivotal role, and fail to function properly in diabetic conditions. Here, we explored the putative contribution of Lp(a) from patients with T2DM with/without DR and healthy controls on inflammation and angiogenesis of retinal endothelial cells (RECs), and on PAC differentiation. Subsequently, we compared the lipid composition of Lp(a) from patients to that from healthy controls., Methods: Lp(a)/LDL obtained from patients and healthy controls were added to TNF-alpha-activated RECs. Expression of VCAM-1/ICAM-1 was measured using flowcytometry. Angiogenesis was determined in REC-pericyte co-cultures stimulated by pro-angiogenic growth factors. PAC differentiation from peripheral blood mononuclear cells was determined by measuring expression of PAC markers. The lipoprotein lipid composition was quantified using detailed lipidomics analysis., Results: Lp(a) from patients with DR (DR-Lp(a)) failed to block TNF-alpha-induced expression of VCAM-1/ICAM-1 in REC whereas Lp(a) from healthy controls (healthy control [HC]-Lp(a)) did. DR-Lp(a) increased REC angiogenesis more than HC-Lp(a) did. Lp(a) from patients without DR showed intermediate profiles. HC-Lp(a) reduced the expression of CD16 and CD105 in PAC, but T2DM-Lp(a) did not. Phosphatidylethanolamine content was lower in T2DM-Lp(a) than in HC-Lp(a)., Conclusions: DR-Lp(a) does not show the anti-inflammatory capacity seen with HC-Lp(a), but increases REC angiogenesis, and affects PAC differentiation less than HC-Lp(a). These functional differences in Lp(a) in T2DM-related retinopathy are associated with alterations in the lipid composition as compared to healthy conditions.
- Published
- 2023
- Full Text
- View/download PDF
21. Pharmacological induction of membrane lipid poly-unsaturation sensitizes melanoma to ROS inducers and overcomes acquired resistance to targeted therapy.
- Author
-
Talebi A, de Laat V, Spotbeen X, Dehairs J, Rambow F, Rogiers A, Vanderhoydonc F, Rizotto L, Planque M, Doglioni G, Motamedi S, Nittner D, Roskams T, Agostinis P, Bechter O, Boecxstaens V, Garmyn M, O'Farrell M, Wagman A, Kemble G, Leucci E, Fendt SM, Marine JC, and Swinnen JV
- Subjects
- Humans, Reactive Oxygen Species metabolism, Membrane Lipids pharmacology, Membrane Lipids therapeutic use, Protein Kinase Inhibitors pharmacology, Cell Line, Tumor, Drug Resistance, Neoplasm, Proto-Oncogene Proteins B-raf genetics, Melanoma drug therapy, Melanoma genetics, Melanoma pathology
- Abstract
Background: One of the key limitations of targeted cancer therapies is the rapid onset of therapy resistance. Taking BRAF-mutant melanoma as paradigm, we previously identified the lipogenic regulator SREBP-1 as a central mediator of resistance to MAPK-targeted therapy. Reasoning that lipogenesis-mediated alterations in membrane lipid poly-unsaturation lie at the basis of therapy resistance, we targeted fatty acid synthase (FASN) as key player in this pathway to evoke an exquisite vulnerability to clinical inducers of reactive oxygen species (ROS), thereby rationalizing a novel clinically actionable combination therapy to overcome therapy resistance., Methods: Using gene expression analysis and mass spectrometry-based lipidomics of BRAF-mutant melanoma cell lines, melanoma PDX and clinical data sets, we explored the association of FASN expression with membrane lipid poly-unsaturation and therapy-resistance. Next, we treated therapy-resistant models with a preclinical FASN inhibitor TVB-3664 and a panel of ROS inducers and performed ROS analysis, lipid peroxidation tests and real-time cell proliferation assays. Finally, we explored the combination of MAPK inhibitors, TVB-3664 and arsenic trioxide (ATO, as a clinically used ROS-inducer) in Mel006 BRAF mutant PDX as a gold model of therapy resistance and assessed the effect on tumor growth, survival and systemic toxicity., Results: We found that FASN expression is consistently increased upon the onset of therapy resistance in clinical melanoma samples, in cell lines and in Mel006 PDX and is associated with decreased lipid poly-unsaturation. Forcing lipid poly-unsaturation in therapy-resistant models by combining MAPK inhibition with FASN inhibition attenuated cell proliferation and rendered cells exquisitely sensitive to a host of ROS inducers. In particular, the triple combination of MAPK inhibition, FASN inhibition, and the clinical ROS-inducing compound ATO dramatically increased survival of Mel006 PDX models from 15 to 72% with no associated signs of toxicity., Conclusions: We conclude that under MAPK inhibition the direct pharmacological inhibition of FASN evokes an exquisite vulnerability to inducers of ROS by increasing membrane lipid poly-unsaturation. The exploitation of this vulnerability by combining MAPK and/or FASN inhibitors with inducers of ROS greatly delays the onset of therapy resistance and increases survival. Our work identifies a clinically actionable combinatorial treatment for therapy-resistant cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
22. The ApoA-I mimetic peptide 5A enhances remyelination by promoting clearance and degradation of myelin debris.
- Author
-
Vanherle S, Jorissen W, Dierckx T, Loix M, Grajchen E, Mingneau F, Guns J, Gervois P, Lambrichts I, Dehairs J, Swinnen JV, Mulder MT, Remaley AT, Haidar M, Hendriks JJA, and Bogie JJF
- Subjects
- Humans, Myelin Sheath metabolism, Apolipoprotein A-I metabolism, Peptides metabolism, Remyelination physiology, Demyelinating Diseases metabolism
- Abstract
The progressive nature of demyelinating diseases lies in the inability of the central nervous system (CNS) to induce proper remyelination. Recently, we and others demonstrated that a dysregulated innate immune response partially underlies failure of CNS remyelination. Extensive accumulation of myelin-derived lipids and an inability to process these lipids was found to induce a disease-promoting phagocyte phenotype. Hence, restoring the ability of these phagocytes to metabolize and efflux myelin-derived lipids represents a promising strategy to promote remyelination. Here, we show that ApoA-I mimetic peptide 5A, a molecule well known to promote activity of the lipid efflux transporter ABCA1, markedly enhances remyelination. Mechanistically, we find that the repair-inducing properties of 5A are attributable to increased clearance and metabolism of remyelination-inhibiting myelin debris via the fatty acid translocase protein CD36, which is transcriptionally controlled by the ABCA1-JAK2-STAT3 signaling pathway. Altogether, our findings indicate that 5A promotes remyelination by stimulating clearance and degradation of myelin debris., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. ORP5/8 and MIB/MICOS link ER-mitochondria and intra-mitochondrial contacts for non-vesicular transport of phosphatidylserine.
- Author
-
Monteiro-Cardoso VF, Rochin L, Arora A, Houcine A, Jääskeläinen E, Kivelä AM, Sauvanet C, Le Bars R, Marien E, Dehairs J, Neveu J, El Khallouki N, Santonico E, Swinnen JV, Tareste D, Olkkonen VM, and Giordano F
- Subjects
- Endoplasmic Reticulum metabolism, Mitochondria metabolism, Mitochondrial Membranes metabolism, Mitochondrial Proteins metabolism, Phosphatidylserines metabolism
- Abstract
Mitochondria are dynamic organelles essential for cell survival whose structural and functional integrity rely on selective and regulated transport of lipids from/to the endoplasmic reticulum (ER) and across the mitochondrial intermembrane space. As they are not connected by vesicular transport, the exchange of lipids between ER and mitochondria occurs at membrane contact sites. However, the mechanisms and proteins involved in these processes are only beginning to emerge. Here, we show that the main physiological localization of the lipid transfer proteins ORP5 and ORP8 is at mitochondria-associated ER membrane (MAM) subdomains, physically linked to the mitochondrial intermembrane space bridging (MIB)/mitochondrial contact sites and cristae junction organizing system (MICOS) complexes that bridge the two mitochondrial membranes. We also show that ORP5/ORP8 mediate non-vesicular transport of phosphatidylserine (PS) lipids from the ER to mitochondria by cooperating with the MIB/MICOS complexes. Overall our study reveals a physical and functional link between ER-mitochondria contacts involved in lipid transfer and intra-mitochondrial membrane contacts maintained by the MIB/MICOS complexes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Perilipin-2 limits remyelination by preventing lipid droplet degradation.
- Author
-
Loix M, Wouters E, Vanherle S, Dehairs J, McManaman JL, Kemps H, Swinnen JV, Haidar M, Bogie JFJ, and Hendriks JJA
- Subjects
- Lipids, Myelin Sheath metabolism, Perilipin-2 genetics, Perilipin-2 metabolism, Lipid Droplets metabolism, Remyelination
- Abstract
Foamy macrophages and microglia containing lipid droplets (LDs) are a pathological hallmark of demyelinating disorders affecting the central nervous system (CNS). We and others showed that excessive accumulation of intracellular lipids drives these phagocytes towards a more inflammatory phenotype, thereby limiting CNS repair. To date, however, the mechanisms underlying LD biogenesis and breakdown in lipid-engorged phagocytes in the CNS, as well as their impact on foamy phagocyte biology and lesion progression, remain poorly understood. Here, we provide evidence that LD-associated protein perilipin-2 (PLIN2) controls LD metabolism in myelin-containing phagocytes. We show that PLIN2 protects LDs from lipolysis-mediated degradation, thereby impairing intracellular processing of myelin-derived lipids in phagocytes. Accordingly, loss of Plin2 stimulates LD turnover in foamy phagocytes, driving them towards a less inflammatory phenotype. Importantly, Plin2-deficiency markedly improves remyelination in the ex vivo brain slice model and in the in vivo cuprizone-induced demyelination model. In summary, we identify PLIN2 as a novel therapeutic target to prevent the pathogenic accumulation of LDs in foamy phagocytes and to stimulate remyelination., (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
25. FTY720 decreases ceramides levels in the brain and prevents memory impairments in a mouse model of familial Alzheimer's disease expressing APOE4.
- Author
-
Crivelli SM, Luo Q, Kruining DV, Giovagnoni C, Mané-Damas M, den Hoedt S, Berkes D, De Vries HE, Mulder MT, Walter J, Waelkens E, Derua R, Swinnen JV, Dehairs J, Wijnands EPM, Bieberich E, Losen M, and Martinez-Martinez P
- Subjects
- Animals, Brain metabolism, Ceramides metabolism, Disease Models, Animal, Fingolimod Hydrochloride metabolism, Fingolimod Hydrochloride pharmacology, Fingolimod Hydrochloride therapeutic use, Humans, Memory Disorders drug therapy, Memory Disorders metabolism, Memory Disorders prevention & control, Mice, Sphingolipids metabolism, Alzheimer Disease drug therapy, Alzheimer Disease genetics, Alzheimer Disease prevention & control, Apolipoprotein E4 genetics, Apolipoprotein E4 metabolism, Apolipoprotein E4 therapeutic use
- Abstract
The protection mediated by the bioactive sphingolipid sphingosine-1-phosphate (S1P) declines during Alzheimer's disease (AD) progression, especially in patients carrying the apolipoprotein E ε4 (APOE4) isoform. The drug FTY720 mimics S1P bioactivity, but its efficacy in treating AD is unclear. Two doses of FTY720 (0.1 mg / kg and 0.5 mg / kg daily) were given by oral gavage for 15 weeks to transgenic mouse models of familial AD carrying human apolipoprotein E (APOE) APOE3 (E3FAD) or APOE4 (E4FAD). After 12 weeks of treatment, animals were subjected to behavioral tests for memory, locomotion, and anxiety. Blood was withdrawn at different time points and brains were collected for sphingolipids analysis by mass spectrometry, gene expression by RT-PCR and Aβ quantification by ELISA. We discovered that low levels of S1P in the plasma is associated with a higher probability of failing the memory test and that FTY720 prevents memory impairments in E4FAD. The beneficial effect of FTY720 was induced by a shift of the sphingolipid metabolism in the brain towards a lower production of toxic metabolites, like ceramide d18:1/16:0 and d18:1/22:0, and reduction of amyloid-β burden and inflammation. In conclusion, we provide further evidence of the druggability of the sphingolipid system in AD., (Copyright © 2022. Published by Elsevier Masson SAS.)
- Published
- 2022
- Full Text
- View/download PDF
26. Deciphering the Role of Extracellular Vesicles Derived from ZIKV-Infected hcMEC/D3 Cells on the Blood-Brain Barrier System.
- Author
-
Fikatas A, Dehairs J, Noppen S, Doijen J, Vanderhoydonc F, Meyen E, Swinnen JV, Pannecouque C, and Schols D
- Subjects
- Blood-Brain Barrier virology, Cells, Cultured, Central Nervous System virology, Endothelial Cells virology, Extracellular Vesicles virology, Humans, Lipidomics, RNA, Viral metabolism, Viral Nonstructural Proteins metabolism, Viral Proteins genetics, Viral Proteins metabolism, Zika Virus Infection virology, Blood-Brain Barrier metabolism, Extracellular Vesicles metabolism, Zika Virus physiology, Zika Virus Infection transmission
- Abstract
To date, no vaccines or antivirals are available against Zika virus (ZIKV). In addition, the mechanisms underlying ZIKV-associated pathogenesis of the central nervous system (CNS) are largely unexplored. Getting more insight into the cellular pathways that ZIKV recruits to facilitate infection of susceptible cells will be crucial for establishing an effective treatment strategy. In general, cells secrete a number of vesicles, known as extracellular vesicles (EVs), in response to viral infections. These EVs serve as intercellular communicators. Here, we investigated the role of EVs derived from ZIKV-infected human brain microvascular endothelial cells on the blood-brain barrier (BBB) system. We demonstrated that ZIKV-infected EVs (IEVs) can incorporate viral components, including ZIKV RNA, NS1, and E-protein, and further transfer them to several types of CNS cells. Using label-free impedance-based biosensing, we observed that ZIKV and IEVs can temporally disturb the monolayer integrity of BBB-mimicking cells, possibly by inducing structural rearrangements of the adherent protein VE-cadherin (immunofluorescence staining). Finally, differences in the lipidomic profile between EVs and their parental cells possibly suggest a preferential sorting mechanism of specific lipid species into the vesicles. To conclude, these data suggest that IEVs could be postulated as vehicles (Trojan horse) for ZIKV transmission via the BBB.
- Published
- 2021
- Full Text
- View/download PDF
27. Regulatory T cell differentiation is controlled by αKG-induced alterations in mitochondrial metabolism and lipid homeostasis.
- Author
-
Matias MI, Yong CS, Foroushani A, Goldsmith C, Mongellaz C, Sezgin E, Levental KR, Talebi A, Perrault J, Rivière A, Dehairs J, Delos O, Bertand-Michel J, Portais JC, Wong M, Marie JC, Kelekar A, Kinet S, Zimmermann VS, Levental I, Yvan-Charvet L, Swinnen JV, Muljo SA, Hernandez-Vargas H, Tardito S, Taylor N, and Dardalhon V
- Subjects
- Animals, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Diacylglycerol O-Acyltransferase metabolism, Fibrosarcoma genetics, Fibrosarcoma immunology, Fibrosarcoma metabolism, Fibrosarcoma therapy, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Homeostasis, Humans, Immunotherapy, Adoptive, Mice, Inbred C57BL, Mice, Knockout, Mitochondria genetics, Mitochondria metabolism, Phenotype, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Signal Transduction, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, T-Lymphocytes, Regulatory transplantation, Th1 Cells drug effects, Th1 Cells immunology, Th1 Cells metabolism, Mice, Cell Differentiation drug effects, Energy Metabolism drug effects, Ketoglutaric Acids pharmacology, Lipid Metabolism drug effects, Mitochondria drug effects, T-Lymphocytes, Regulatory drug effects
- Abstract
Suppressive regulatory T cell (Treg) differentiation is controlled by diverse immunometabolic signaling pathways and intracellular metabolites. Here we show that cell-permeable α-ketoglutarate (αKG) alters the DNA methylation profile of naive CD4 T cells activated under Treg polarizing conditions, markedly attenuating FoxP3+ Treg differentiation and increasing inflammatory cytokines. Adoptive transfer of these T cells into tumor-bearing mice results in enhanced tumor infiltration, decreased FoxP3 expression, and delayed tumor growth. Mechanistically, αKG leads to an energetic state that is reprogrammed toward a mitochondrial metabolism, with increased oxidative phosphorylation and expression of mitochondrial complex enzymes. Furthermore, carbons from ectopic αKG are directly utilized in the generation of fatty acids, associated with lipidome remodeling and increased triacylglyceride stores. Notably, inhibition of either mitochondrial complex II or DGAT2-mediated triacylglyceride synthesis restores Treg differentiation and decreases the αKG-induced inflammatory phenotype. Thus, we identify a crosstalk between αKG, mitochondrial metabolism and triacylglyceride synthesis that controls Treg fate., Competing Interests: Declaration of interests C.M., S.K., V.D., and N.T. are inventors on patents describing the use of ligands for detection of and modulation of metabolite transporters (N.T. gave up her rights), licensed to METAFORA-biosystems., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
28. Lipidomic Profiling of Clinical Prostate Cancer Reveals Targetable Alterations in Membrane Lipid Composition.
- Author
-
Butler LM, Mah CY, Machiels J, Vincent AD, Irani S, Mutuku SM, Spotbeen X, Bagadi M, Waltregny D, Moldovan M, Dehairs J, Vanderhoydonc F, Bloch K, Das R, Stahl J, Kench JG, Gevaert T, Derua R, Waelkens E, Nassar ZD, Selth LA, Trim PJ, Snel MF, Lynn DJ, Tilley WD, Horvath LG, Centenera MM, and Swinnen JV
- Subjects
- Biomarkers, Computational Biology methods, Energy Metabolism, Humans, Male, Metabolomics methods, Molecular Targeted Therapy, Neoplasm Grading, Neoplasm Staging, Prostatic Neoplasms diagnosis, Prostatic Neoplasms drug therapy, Prostatic Neoplasms etiology, Spectrometry, Mass, Electrospray Ionization, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Tandem Mass Spectrometry, Transcriptional Regulator ERG genetics, Transcriptional Regulator ERG metabolism, Lipid Metabolism drug effects, Lipidomics methods, Membrane Lipids metabolism, Prostatic Neoplasms metabolism
- Abstract
Dysregulated lipid metabolism is a prominent feature of prostate cancer that is driven by androgen receptor (AR) signaling. Here we used quantitative mass spectrometry to define the "lipidome" in prostate tumors with matched benign tissues ( n = 21), independent unmatched tissues ( n = 47), and primary prostate explants cultured with the clinical AR antagonist enzalutamide ( n = 43). Significant differences in lipid composition were detected and spatially visualized in tumors compared with matched benign samples. Notably, tumors featured higher proportions of monounsaturated lipids overall and elongated fatty acid chains in phosphatidylinositol and phosphatidylserine lipids. Significant associations between lipid profile and malignancy were validated in unmatched samples, and phospholipid composition was characteristically altered in patient tissues that responded to AR inhibition. Importantly, targeting tumor-related lipid features via inhibition of acetyl-CoA carboxylase 1 significantly reduced cellular proliferation and induced apoptosis in tissue explants. This characterization of the prostate cancer lipidome in clinical tissues reveals enhanced fatty acid synthesis, elongation, and desaturation as tumor-defining features, with potential for therapeutic targeting. SIGNIFICANCE: This study identifies malignancy and treatment-associated changes in lipid composition of clinical prostate cancer tissues, suggesting that mediators of these lipidomic changes could be targeted using existing metabolic agents., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. New Insights on the PBMCs Phospholipidome in Obesity Demonstrate Modulations Associated with Insulin Resistance and Glycemic Status.
- Author
-
Wilkin C, Colonval M, Dehairs J, Esser N, Iovino M, Gianfrancesco MA, Fadeur M, Swinnen JV, Paquot N, Piette J, and Legrand-Poels S
- Subjects
- Adult, Biomarkers, Body Weights and Measures, Computational Biology, Diabetes Mellitus, Type 2 etiology, Diabetes Mellitus, Type 2 metabolism, Female, Glycated Hemoglobin metabolism, Humans, Male, Mass Spectrometry, Membrane Lipids, Middle Aged, Obesity blood, Obesity etiology, Young Adult, Blood Glucose, Insulin Resistance, Leukocytes, Mononuclear metabolism, Lipidomics methods, Obesity metabolism, Phospholipids metabolism
- Abstract
(1) Background: Obesity and type 2 diabetes have been suspected to impact both intrinsic metabolism and function of circulating immune cells. (2) Methods: To further investigate this immunometabolic modulation, we profiled the phospholipidome of the peripheral blood mononuclear cells (PBMCs) in lean, normoglycemic obese (OBNG) and obese with dysglycemia (OBDysG) individuals. (3) Results: The global PBMCs phospholipidome is significantly downmodulated in OBDysG unlike OBNG patients when compared to lean ones. Multiple linear regression analyses show a strong negative relationship between the global PBMCs phospholipidome and parameters assessing insulin resistance. Even though all classes of phospholipid are affected, the relative abundance of each class is maintained with the exception of Lyso-PC/PC and Lyso-PE/PE ratios that are downmodulated in PBMCs of OBDysG compared to OBNG individuals. Interestingly, the percentage of saturated PC is positively associated with glycated hemoglobin (HbA1c). Moreover, a few lipid species are significantly downmodulated in PBMCs of OBDysG compared to OBNG individuals, making possible to distinguish the two phenotypes. (4) Conclusions: This lipidomic study highlights for the first-time modulations of the PBMCs phospholipidome in obese patients with prediabetes and type 2 diabetes. Such phospholipidome remodeling could disrupt the cell membranes and the lipid mediator's levels, driving an immune cell dysfunction.
- Published
- 2021
- Full Text
- View/download PDF
30. Fat Induces Glucose Metabolism in Nontransformed Liver Cells and Promotes Liver Tumorigenesis.
- Author
-
Broadfield LA, Duarte JAG, Schmieder R, Broekaert D, Veys K, Planque M, Vriens K, Karasawa Y, Napolitano F, Fujita S, Fujii M, Eto M, Holvoet B, Vangoitsenhoven R, Fernandez-Garcia J, Van Elsen J, Dehairs J, Zeng J, Dooley J, Rubio RA, van Pelt J, Grünewald TGP, Liston A, Mathieu C, Deroose CM, Swinnen JV, Lambrechts D, di Bernardo D, Kuroda S, De Bock K, and Fendt SM
- Subjects
- Animals, Carcinoma, Hepatocellular metabolism, Cell Transformation, Neoplastic, Citric Acid Cycle physiology, Fatty Acids metabolism, Glucose Tolerance Test, Humans, Lactic Acid biosynthesis, Lipid Metabolism, Liver Neoplasms metabolism, Mice, Mice, Inbred C57BL, Obesity complications, Palmitates pharmacology, Peroxisomes metabolism, Proteomics, Pyruvate Carboxylase metabolism, Random Allocation, Reactive Oxygen Species metabolism, Serine metabolism, Transcriptional Activation, Carcinoma, Hepatocellular etiology, Diet, High-Fat, Dietary Fats metabolism, Glucose metabolism, Hepatocytes metabolism, Liver Neoplasms etiology
- Abstract
Hepatic fat accumulation is associated with diabetes and hepatocellular carcinoma (HCC). Here, we characterize the metabolic response that high-fat availability elicits in livers before disease development. After a short term on a high-fat diet (HFD), otherwise healthy mice showed elevated hepatic glucose uptake and increased glucose contribution to serine and pyruvate carboxylase activity compared with control diet (CD) mice. This glucose phenotype occurred independently from transcriptional or proteomic programming, which identifies increased peroxisomal and lipid metabolism pathways. HFD-fed mice exhibited increased lactate production when challenged with glucose. Consistently, administration of an oral glucose bolus to healthy individuals revealed a correlation between waist circumference and lactate secretion in a human cohort. In vitro , palmitate exposure stimulated production of reactive oxygen species and subsequent glucose uptake and lactate secretion in hepatocytes and liver cancer cells. Furthermore, HFD enhanced the formation of HCC compared with CD in mice exposed to a hepatic carcinogen. Regardless of the dietary background, all murine tumors showed similar alterations in glucose metabolism to those identified in fat exposed nontransformed mouse livers, however, particular lipid species were elevated in HFD tumor and nontumor-bearing HFD liver tissue. These findings suggest that fat can induce glucose-mediated metabolic changes in nontransformed liver cells similar to those found in HCC. SIGNIFICANCE: With obesity-induced hepatocellular carcinoma on a rising trend, this study shows in normal, nontransformed livers that fat induces glucose metabolism similar to an oncogenic transformation., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
31. ELOVL5 Is a Critical and Targetable Fatty Acid Elongase in Prostate Cancer.
- Author
-
Centenera MM, Scott JS, Machiels J, Nassar ZD, Miller DC, Zinonos I, Dehairs J, Burvenich IJG, Zadra G, Chetta PM, Bango C, Evergren E, Ryan NK, Gillis JL, Mah CY, Tieu T, Hanson AR, Carelli R, Bloch K, Panagopoulos V, Waelkens E, Derua R, Williams ED, Evdokiou A, Cifuentes-Rius A, Voelcker NH, Mills IG, Tilley WD, Scott AM, Loda M, Selth LA, Swinnen JV, and Butler LM
- Subjects
- Animals, Cell Movement drug effects, Cell Movement genetics, Cell Proliferation drug effects, Cell Proliferation genetics, Fatty Acid Elongases genetics, Fatty Acid Elongases physiology, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, Humans, Lipid Metabolism genetics, Male, Mice, Mice, Inbred NOD, Mice, SCID, Molecular Targeted Therapy methods, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, RNA, Small Interfering pharmacology, Receptors, Androgen physiology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Fatty Acid Elongases antagonists & inhibitors, Prostatic Neoplasms drug therapy, RNA, Small Interfering therapeutic use
- Abstract
The androgen receptor (AR) is the key oncogenic driver of prostate cancer, and despite implementation of novel AR targeting therapies, outcomes for metastatic disease remain dismal. There is an urgent need to better understand androgen-regulated cellular processes to more effectively target the AR dependence of prostate cancer cells through new therapeutic vulnerabilities. Transcriptomic studies have consistently identified lipid metabolism as a hallmark of enhanced AR signaling in prostate cancer, yet the relationship between AR and the lipidome remains undefined. Using mass spectrometry-based lipidomics, this study reveals increased fatty acyl chain length in phospholipids from prostate cancer cells and patient-derived explants as one of the most striking androgen-regulated changes to lipid metabolism. Potent and direct AR-mediated induction of ELOVL fatty acid elongase 5 (ELOVL5), an enzyme that catalyzes fatty acid elongation, was demonstrated in prostate cancer cells, xenografts, and clinical tumors. Assessment of mRNA and protein in large-scale data sets revealed ELOVL5 as the predominant ELOVL expressed and upregulated in prostate cancer compared with nonmalignant prostate. ELOVL5 depletion markedly altered mitochondrial morphology and function, leading to excess generation of reactive oxygen species and resulting in suppression of prostate cancer cell proliferation, 3D growth, and in vivo tumor growth and metastasis. Supplementation with the monounsaturated fatty acid cis-vaccenic acid, a direct product of ELOVL5 elongation, reversed the oxidative stress and associated cell proliferation and migration effects of ELOVL5 knockdown. Collectively, these results identify lipid elongation as a protumorigenic metabolic pathway in prostate cancer that is androgen-regulated, critical for metastasis, and targetable via ELOVL5. SIGNIFICANCE: This study identifies phospholipid elongation as a new metabolic target of androgen action that is critical for prostate tumor metastasis., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
32. Synthesis and fluorine-18 radiolabeling of a phospholipid as a PET imaging agent for prostate cancer.
- Author
-
Kwan KH, Burvenich IJG, Centenera MM, Goh YW, Rigopoulos A, Dehairs J, Swinnen JV, Raj GV, Hoy AJ, Butler LM, Scott AM, White JM, and Ackermann U
- Subjects
- Male, Humans, Animals, Cell Line, Tumor, Tissue Distribution, Mice, Chemistry Techniques, Synthetic, Radiochemistry, Phospholipids chemistry, Phospholipids chemical synthesis, Phospholipids pharmacokinetics, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms metabolism, Fluorine Radioisotopes chemistry, Positron-Emission Tomography methods, Isotope Labeling
- Abstract
Introduction: Altered lipid metabolism and subsequent changes in cellular lipid composition have been observed in prostate cancer cells, are associated with poor clinical outcome, and are promising targets for metabolic therapies. This study reports for the first time on the synthesis of a phospholipid radiotracer based on the phospholipid 1,2-didocosahexaenoyl-sn-glycero-3-phosphocholine (PC44:12) to allow tracking of polyunsaturated lipid tumor uptake via PET imaging. This tracer may aid in the development of strategies to modulate response to therapies targeting lipid metabolism in prostate cancer., Methods: Lipidomics analysis of prostate tumor explants and LNCaP tumor cells were used to identify PC44:12 as a potential phospholipid candidate for radiotracer development. Synthesis of phosphocholine precursor and non-radioactive standard were optimised using click chemistry. The biodistribution of a fluorine-18 labeled analogue, N-{[4-(2-[
18 F]fluoroethyl)-2,3,4-triazol-1-yl]methyl}-1,2-didocosahexaenoyl-sn-glycero-3-phosphocholine ([18 F]2) was determined in LNCaP prostate tumor-bearing NOD SCID gamma mice by ex vivo biodistribution and PET imaging studies and compared to biodistribution of [18 F]fluoromethylcholine., Results: [18 F]2 was produced with a decay-corrected yield of 17.8 ± 3.7% and an average radiochemical purity of 97.00 ± 0.89% (n = 6). Molar activity was 85.1 ± 3.45 GBq/μmol (2300 ± 93 mCi/μmol) and the total synthesis time was 2 h. Ex vivo biodistribution data demonstrated high liver uptake (41.1 ± 9.2%ID/g) and high splenic uptake (10.9 ± 9.1%ID/g) 50 min post-injection. Ex vivo biodistribution showed low absolute tumor uptake of [18 F]2 (0.8 ± 0.3%ID/g). However, dynamic PET imaging demonstrated an increase over time of the relative tumor-to-muscle ratio with a peak of 2.8 ± 0.5 reached 1 h post-injection. In contrast, dynamic PET of [18 F]fluoromethylcholine demonstrated no increase in tumor-to-muscle ratios due to an increase in both tumor and muscle over time. Absolute uptake of [18 F]fluoromethylcholine was higher and peaked at 60 min post injection (2.25 ± 0.29%ID/g) compared to [18 F]2 (1.44 ± 0.06%ID/g) during the 1 h dynamic scan period., Conclusions and Advances in Knowledge: This study demonstrates the ability to radiolabel phospholipids and indicates the potential to monitor the in vivo distribution of phospholipids using fluorine-18 based PET., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
33. Ceramide analog [ 18 F]F-HPA-12 detects sphingolipid disbalance in the brain of Alzheimer's disease transgenic mice by functioning as a metabolic probe.
- Author
-
Crivelli SM, van Kruining D, Luo Q, Stevens JAA, Giovagnoni C, Paulus A, Bauwens M, Berkes D, de Vries HE, Mulder MT, Walter J, Waelkens E, Derua R, Swinnen JV, Dehairs J, Mottaghy FM, Losen M, Bieberich E, and Martinez-Martinez P
- Subjects
- Alzheimer Disease diagnostic imaging, Animals, Apolipoprotein E3 genetics, Apolipoprotein E4 genetics, Astrocytes metabolism, Brain diagnostic imaging, Disease Models, Animal, Female, Hippocampus metabolism, Lipidomics, Mass Spectrometry, Mice, Mice, Knockout, ApoE, ROC Curve, Sensitivity and Specificity, Sphingomyelins metabolism, Alzheimer Disease genetics, Amides, Brain metabolism, Ceramides chemistry, Fluorine Radioisotopes, Sphingolipids chemistry
- Abstract
The metabolism of ceramides is deregulated in the brain of Alzheimer's disease (AD) patients and is associated with apolipoprotein (APO) APOE4 and amyloid-β pathology. However, how the ceramide metabolism changes over time in AD, in vivo, remains unknown. Distribution and metabolism of [
18 F]F-HPA-12, a radio-fluorinated version of the ceramide analog N-(3-hydroxy-1-hydroxymethyl-3-phenylpropyl) dodecanamide, was investigated in the brain of AD transgenic mouse models (FAD) on an APOE4 or APOE3 genetic background, by positron emission tomography and by gamma counter. We found that FAD mice displayed a higher uptake of [18 F]F-HPA-12 in the brain, independently from the APOE4 or APOE3 genetic background. FAD mice could be distinguished from littermate control animals with a sensitivity of 85.7% and a specificity of 87.5%, by gamma counter measurements. Metabolic analysis of [18 F]F-HPA-12 in the brain suggested that the tracer is degraded less efficiently in the FAD mice. Furthermore, the radioactive signal registered in the hippocampus correlated with an increase of Cer d18:1/20:2 levels measured in the same brain region by mass spectrometry. Our data gives additional proof that ceramide metabolism is different in FAD mice compared to controls. Ceramide analogs like HPA-12 may function as metabolic probes to study ceramide disbalance in the brain.- Published
- 2020
- Full Text
- View/download PDF
34. The role of lipids in determining the air-water interfacial properties of wheat, rye, and oat dough liquor constituents.
- Author
-
Janssen F, Wouters AGB, Linclau L, Waelkens E, Derua R, Dehairs J, Moldenaers P, Vermant J, and Delcour JA
- Subjects
- Bread analysis, Phase Transition, Surface Tension, Viscosity, Water chemistry, Avena chemistry, Lipids chemistry, Secale chemistry, Triticum chemistry
- Abstract
Bread is mainly made from wheat but also from other cereals such as rye and oats. We here report on the role of dough liquor (DL) proteins and lipids in determining the stability of gas cell air-water (A-W) interfaces in wheat, rye, and oat bread making. Surprisingly, most lipids in DLs of these cereals are nonpolar. Their main polar DL lipids are phospholipids. Lipids at wheat and rye DL stabilized A-W interfaces impair interactions between its proteins, as reflected by an increased A-W interfacial shear viscosity of the adsorbed film upon defatting. In contrast, removing most lipids from oat DL pronouncedly increased the A-W interface surface tension, demonstrating that lipids are the prominent adsorbed species., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis.
- Author
-
Nassar ZD, Mah CY, Dehairs J, Burvenich IJ, Irani S, Centenera MM, Helm M, Shrestha RK, Moldovan M, Don AS, Holst J, Scott AM, Horvath LG, Lynn DJ, Selth LA, Hoy AJ, Swinnen JV, and Butler LM
- Subjects
- Cell Line, Tumor, Fatty Acids, Unsaturated metabolism, Humans, Male, Oxidation-Reduction, Oxidoreductases Acting on CH-CH Group Donors metabolism, Prostatic Neoplasms genetics, Ferroptosis, Oxidoreductases Acting on CH-CH Group Donors genetics, Prostatic Neoplasms physiopathology
- Abstract
Fatty acid β-oxidation (FAO) is the main bioenergetic pathway in human prostate cancer (PCa) and a promising novel therapeutic vulnerability. Here we demonstrate therapeutic efficacy of targeting FAO in clinical prostate tumors cultured ex vivo, and identify DECR1, encoding the rate-limiting enzyme for oxidation of polyunsaturated fatty acids (PUFAs), as robustly overexpressed in PCa tissues and associated with shorter relapse-free survival. DECR1 is a negatively-regulated androgen receptor (AR) target gene and, therefore, may promote PCa cell survival and resistance to AR targeting therapeutics. DECR1 knockdown selectively inhibited β-oxidation of PUFAs, inhibited proliferation and migration of PCa cells, including treatment resistant lines, and suppressed tumor cell proliferation and metastasis in mouse xenograft models. Mechanistically, targeting of DECR1 caused cellular accumulation of PUFAs, enhanced mitochondrial oxidative stress and lipid peroxidation, and induced ferroptosis. These findings implicate PUFA oxidation via DECR1 as an unexplored facet of FAO that promotes survival of PCa cells., Competing Interests: ZN, CM, JD, IB, SI, MC, MH, RS, MM, AD, JH, AS, LH, DL, LS, AH, JS, LB No competing interests declared, (© 2020, Nassar et al.)
- Published
- 2020
- Full Text
- View/download PDF
36. Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain.
- Author
-
Bogie JFJ, Grajchen E, Wouters E, Corrales AG, Dierckx T, Vanherle S, Mailleux J, Gervois P, Wolfs E, Dehairs J, Van Broeckhoven J, Bowman AP, Lambrichts I, Gustafsson JÅ, Remaley AT, Mulder M, Swinnen JV, Haidar M, Ellis SR, Ntambi JM, Zelcer N, and Hendriks JJA
- Subjects
- ATP Binding Cassette Transporter 1 metabolism, Animals, Cell Line, Cholesterol metabolism, Endocytosis, Fatty Acids metabolism, Foam Cells metabolism, Humans, Inflammation pathology, Macrophages metabolism, Macrophages ultrastructure, Mice, Microglia metabolism, Myelin Sheath metabolism, Phagocytes pathology, Phagocytes ultrastructure, Phenotype, Protein Kinase C-delta metabolism, Stearoyl-CoA Desaturase deficiency, Brain pathology, Macrophages enzymology, Microglia enzymology, Stearoyl-CoA Desaturase metabolism
- Abstract
Failure of remyelination underlies the progressive nature of demyelinating diseases such as multiple sclerosis. Macrophages and microglia are crucially involved in the formation and repair of demyelinated lesions. Here we show that myelin uptake temporarily skewed these phagocytes toward a disease-resolving phenotype, while sustained intracellular accumulation of myelin induced a lesion-promoting phenotype. This phenotypic shift was controlled by stearoyl-CoA desaturase-1 (SCD1), an enzyme responsible for the desaturation of saturated fatty acids. Monounsaturated fatty acids generated by SCD1 reduced the surface abundance of the cholesterol efflux transporter ABCA1, which in turn promoted lipid accumulation and induced an inflammatory phagocyte phenotype. Pharmacological inhibition or phagocyte-specific deficiency of Scd1 accelerated remyelination ex vivo and in vivo. These findings identify SCD1 as a novel therapeutic target to promote remyelination., Competing Interests: Disclosures: The authors declare no competing interests exist., (© 2020 Bogie et al.)
- Published
- 2020
- Full Text
- View/download PDF
37. Expression Levels of 4 Genes in Colon Tissue Might Be Used to Predict Which Patients Will Enter Endoscopic Remission After Vedolizumab Therapy for Inflammatory Bowel Diseases.
- Author
-
Verstockt B, Verstockt S, Veny M, Dehairs J, Arnauts K, Van Assche G, De Hertogh G, Vermeire S, Salas A, and Ferrante M
- Subjects
- Antibodies, Monoclonal, Humanized, Argonaute Proteins, Colon, Gastrointestinal Agents therapeutic use, Humans, Remission Induction, Treatment Outcome, Tumor Necrosis Factor Inhibitors, Colitis, Ulcerative drug therapy, Inflammatory Bowel Diseases drug therapy, RGS Proteins
- Abstract
Background & Aims: We aimed to identify biomarkers that might be used to predict responses of patients with inflammatory bowel diseases (IBD) to vedolizumab therapy., Methods: We obtained biopsies from inflamed colon of patients with IBD who began treatment with vedolizumab (n = 31) or tumor necrosis factor (TNF) antagonists (n = 20) and performed RNA-sequencing analyses. We compared gene expression patterns between patients who did and did not enter endoscopic remission (absence of ulcerations at month 6 for patients with Crohn's disease or Mayo endoscopic subscore ≤1 at week 14 for patients with ulcerative colitis) and performed pathway analysis and cell deconvolution for training (n = 20) and validation (n = 11) datasets. Colon biopsies were also analyzed by immunohistochemistry. We validated a baseline gene expression pattern associated with endoscopic remission after vedolizumab therapy using 3 independent datasets (n = 66)., Results: We identified significant differences in expression levels of 44 genes between patients who entered remission after vedolizumab and those who did not; we found significant increases in leukocyte migration in colon tissues from patients who did not enter remission (P < .006). Deconvolution methods identified a significant enrichment of monocytes (P = .005), M1-macrophages (P = .05), and CD4+ T cells (P = .008) in colon tissues from patients who did not enter remission, whereas colon tissues from patients in remission had higher numbers of naïve B cells before treatment (P = .05). Baseline expression levels of PIWIL1, MAATS1, RGS13, and DCHS2 identified patients who did vs did not enter remission with 80% accuracy in the training set and 100% accuracy in validation dataset 1. We validated these findings in the 3 independent datasets by microarray, RNA sequencing and quantitative PCR analysis (P = .003). Expression levels of these 4 genes did not associate with response to anti-TNF agents. We confirmed the presence of proteins encoded by mRNAs using immunohistochemistry., Conclusions: We identified 4 genes whose baseline expression levels in colon tissues of patients with IBD associate with endoscopic remission after vedolizumab, but not anti-TNF, treatment. We validated this signature in 4 independent datasets and also at the protein level. Studies of these genes might provide insights into the mechanisms of action of vedolizumab., (Copyright © 2020 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Endocytosis of very low-density lipoproteins: an unexpected mechanism for lipid acquisition by breast cancer cells.
- Author
-
Lupien LE, Bloch K, Dehairs J, Traphagen NA, Feng WW, Davis WL, Dennis T, Swinnen JV, Wells WA, Smits NC, Kuemmerle NB, Miller TW, and Kinlaw WB
- Subjects
- Humans, Tumor Cells, Cultured, Breast Neoplasms metabolism, Breast Neoplasms pathology, Endocytosis, Lipid Droplets metabolism, Lipoproteins, VLDL metabolism
- Abstract
We previously described the expression of CD36 and LPL by breast cancer (BC) cells and tissues and the growth-promoting effect of VLDL observed only in the presence of LPL. We now report a model in which LPL is bound to a heparan sulfate proteoglycan motif on the BC cell surface and acts in concert with the VLDL receptor to internalize VLDLs via receptor-mediated endocytosis. We also demonstrate that gene-expression programs for lipid synthesis versus uptake respond robustly to triglyceride-rich lipoprotein availability. The literature emphasizes de novo FA synthesis and exogenous free FA uptake using CD36 as paramount mechanisms for lipid acquisition by cancer cells. We find that the uptake of intact lipoproteins is also an important mechanism for lipid acquisition and that the relative reliance on lipid synthesis versus uptake varies among BC cell lines and in response to VLDL availability. This metabolic plasticity has important implications for the development of therapies aimed at the lipid dependence of many types of cancer, in that the inhibition of FA synthesis may elicit compensatory upregulation of lipid uptake. Moreover, the mechanism that we have elucidated provides a direct connection between dietary fat and tumor biology.-.
- Published
- 2020
- Full Text
- View/download PDF
39. Lipids and cancer: Emerging roles in pathogenesis, diagnosis and therapeutic intervention.
- Author
-
Butler LM, Perone Y, Dehairs J, Lupien LE, de Laat V, Talebi A, Loda M, Kinlaw WB, and Swinnen JV
- Subjects
- Animals, Biomarkers metabolism, Humans, Lipids, Neoplasms diagnosis, Neoplasms therapy, Lipid Metabolism, Neoplasms metabolism
- Abstract
With the advent of effective tools to study lipids, including mass spectrometry-based lipidomics, lipids are emerging as central players in cancer biology. Lipids function as essential building blocks for membranes, serve as fuel to drive energy-demanding processes and play a key role as signaling molecules and as regulators of numerous cellular functions. Not unexpectedly, cancer cells, as well as other cell types in the tumor microenvironment, exploit various ways to acquire lipids and extensively rewire their metabolism as part of a plastic and context-dependent metabolic reprogramming that is driven by both oncogenic and environmental cues. The resulting changes in the fate and composition of lipids help cancer cells to thrive in a changing microenvironment by supporting key oncogenic functions and cancer hallmarks, including cellular energetics, promoting feedforward oncogenic signaling, resisting oxidative and other stresses, regulating intercellular communication and immune responses. Supported by the close connection between altered lipid metabolism and the pathogenic process, specific lipid profiles are emerging as unique disease biomarkers, with diagnostic, prognostic and predictive potential. Multiple preclinical studies illustrate the translational promise of exploiting lipid metabolism in cancer, and critically, have shown context dependent actionable vulnerabilities that can be rationally targeted, particularly in combinatorial approaches. Moreover, lipids themselves can be used as membrane disrupting agents or as key components of nanocarriers of various therapeutics. With a number of preclinical compounds and strategies that are approaching clinical trials, we are at the doorstep of exploiting a hitherto underappreciated hallmark of cancer and promising target in the oncologist's strategy to combat cancer., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. The Transfer of Sphingomyelinase Contributes to Drug Resistance in Multiple Myeloma.
- Author
-
Faict S, Oudaert I, D'Auria L, Dehairs J, Maes K, Vlummens P, De Veirman K, De Bruyne E, Fostier K, Vande Broek I, Schots R, Vanderkerken K, Swinnen JV, and Menu E
- Abstract
Multiple myeloma (MM) is well-known for the development of drug resistance, leading to relapse. Therefore, finding novel treatment strategies remains necessary. By performing a lipidomics assay on MM patient plasma, we aimed to identify new targets. We observed a dysregulation in the sphingolipid metabolism, with the upregulation of several ceramides and downregulation of sphingomyelin. This imbalance suggests an increase in sphingomyelinase, the enzyme responsible for hydrolyzing sphingomyelin into ceramide. We confirmed the upregulation of acid sphingomyelinase (ASM) in primary MM cells. Furthermore, we observed an increase in ASM expression in MM cell lines treated with melphalan or bortezomib, as well as in their exosomes. Exosomes high in ASM content were able to transfer the drug-resistant phenotype to chemosensitive cells, hereby suggesting a tumor-protective role for ASM. Finally, inhibition of ASM by amitriptyline improved drug sensitivity in MM cell lines and primary MM cells. In summary, this study is the first to analyze differences in plasma lipid composition of MM patients and match the observed differences to an upregulation of ASM. Moreover, we demonstrate that amitriptyline is able to inhibit ASM and increase sensitivity to anti-myeloma drugs. This study, therefore, provides a rational to include ASM-targeting-drugs in combination strategies in myeloma patients., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
41. A New Classification Method of Metastatic Cancers Using a 1 H-NMR-Based Approach: A Study Case of Melanoma, Breast, and Prostate Cancer Cell Lines.
- Author
-
Schepkens C, Dallons M, Dehairs J, Talebi A, Jeandriens J, Drossart LM, Auquier G, Tagliatti V, Swinnen JV, and Colet JM
- Abstract
In this study, metastatic melanoma, breast, and prostate cancer cell lines were analyzed using a
1 H-NMR-based approach in order to investigate common features and differences of aggressive cancers metabolomes. For that purpose,1 H-NMR spectra of both cellular extracts and culture media were combined with multivariate data analysis, bringing to light no less than 20 discriminant metabolites able to separate the metastatic metabolomes. The supervised approach succeeded in classifying the metastatic cell lines depending on their glucose metabolism, more glycolysis-oriented in the BRAF proto-oncogene mutated cell lines compared to the others. Other adaptive metabolic features also contributed to the classification, such as the increased total choline content (tCho), UDP-GlcNAc detection, and various changes in the glucose-related metabolites tree, giving additional information about the metastatic metabolome status and direction. Finally, common metabolic features detected via1 H-NMR in the studied cancer cell lines are discussed, identifying the glycolytic pathway, Kennedy's pathway, and the glutaminolysis as potential and common targets in metastasis, opening up new avenues to cure cancer., Competing Interests: The authors declare no conflict of interest- Published
- 2019
- Full Text
- View/download PDF
42. Membrane Lipid Remodeling Takes Center Stage in Growth Factor Receptor-Driven Cancer Development.
- Author
-
Swinnen JV, Dehairs J, and Talebi A
- Subjects
- Humans, Intercellular Signaling Peptides and Proteins, Lipid Metabolism, Membrane Lipids, Receptors, Growth Factor, Neoplasms
- Abstract
Enhanced growth factor signaling is a hallmark of cancer, allowing cancer cells to thrive in a challenging environment. In this issue of Cell Metabolism, Bi et al. (2019) identify LPCAT1, a key membrane lipid remodeling enzyme, as a central link between genetically driven growth factor receptor expression, signaling, and tumor growth, highlighting lipid remodeling as a therapeutic target in cancer., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
43. Saturated fatty acids induce NLRP3 activation in human macrophages through K + efflux resulting from phospholipid saturation and Na, K-ATPase disruption.
- Author
-
Gianfrancesco MA, Dehairs J, L'homme L, Herinckx G, Esser N, Jansen O, Habraken Y, Lassence C, Swinnen JV, Rider MH, Piette J, Paquot N, and Legrand-Poels S
- Subjects
- Biological Transport, Cells, Cultured, Humans, Inflammasomes metabolism, Phospholipids metabolism, Fatty Acids metabolism, Macrophages metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Potassium metabolism, Sodium-Potassium-Exchanging ATPase metabolism
- Abstract
NLRP3 inflammasome plays a key role in Western diet-induced systemic inflammation and was recently shown to mediate long-lasting trained immunity in myeloid cells. Saturated fatty acids (SFAs) are sterile triggers able to induce the assembly of the NLRP3 inflammasome in macrophages, leading to IL-1β secretion while unsaturated ones (UFAs) prevent SFAs-mediated NLRP3 activation. Unlike previous studies using LPS-primed bone marrow derived macrophages, we do not see any ROS or IRE-1α involvement in SFAs-mediated NLRP3 activation in human monocytes-derived macrophages. Rather we show that SFAs need to enter the cells and to be activated into acyl-CoA to lead to NLRP3 activation in human macrophages. However, their β-oxidation is dispensable. Instead, they are channeled towards phospholipids but redirected towards lipid droplets containing triacylglycerol in the presence of UFAs. Lipidomic analyses and Laurdan fluorescence experiments demonstrate that SFAs induce a dramatic saturation of phosphatidylcholine (PC) correlated with a loss of membrane fluidity, both events inhibited by UFAs. The silencing of CCTα, the key enzyme in PC synthesis, prevents SFA-mediated NLRP3 activation, demonstrating the essential role of the de novo PC synthesis. This SFA-induced membrane remodeling promotes a disruption of the plasma membrane Na, K-ATPase, instigating a K
+ efflux essential and sufficient for NLRP3 activation. This work opens novel therapeutic avenues to interfere with Western diet-associated diseases such as those targeting the glycerolipid pathway., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
44. Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity.
- Author
-
Vriens K, Christen S, Parik S, Broekaert D, Yoshinaga K, Talebi A, Dehairs J, Escalona-Noguero C, Schmieder R, Cornfield T, Charlton C, Romero-Pérez L, Rossi M, Rinaldi G, Orth MF, Boon R, Kerstens A, Kwan SY, Faubert B, Méndez-Lucas A, Kopitz CC, Chen T, Fernandez-Garcia J, Duarte JAG, Schmitz AA, Steigemann P, Najimi M, Hägebarth A, Van Ginderachter JA, Sokal E, Gotoh N, Wong KK, Verfaillie C, Derua R, Munck S, Yuneva M, Beretta L, DeBerardinis RJ, Swinnen JV, Hodson L, Cassiman D, Verslype C, Christian S, Grünewald S, Grünewald TGP, and Fendt SM
- Subjects
- Animals, Cell Line, Tumor, Cell Membrane metabolism, Cell Proliferation, Fatty Acid Desaturases metabolism, Female, HEK293 Cells, Humans, Male, Mice, Oleic Acids metabolism, Palmitates metabolism, Palmitic Acids metabolism, Stearoyl-CoA Desaturase metabolism, Fatty Acids chemistry, Fatty Acids metabolism, Metabolic Networks and Pathways, Neoplasms metabolism, Neoplasms pathology
- Abstract
Most tumours have an aberrantly activated lipid metabolism
1,2 that enables them to synthesize, elongate and desaturate fatty acids to support proliferation. However, only particular subsets of cancer cells are sensitive to approaches that target fatty acid metabolism and, in particular, fatty acid desaturation3 . This suggests that many cancer cells contain an unexplored plasticity in their fatty acid metabolism. Here we show that some cancer cells can exploit an alternative fatty acid desaturation pathway. We identify various cancer cell lines, mouse hepatocellular carcinomas, and primary human liver and lung carcinomas that desaturate palmitate to the unusual fatty acid sapienate to support membrane biosynthesis during proliferation. Accordingly, we found that sapienate biosynthesis enables cancer cells to bypass the known fatty acid desaturation pathway that is dependent on stearoyl-CoA desaturase. Thus, only by targeting both desaturation pathways is the in vitro and in vivo proliferation of cancer cells that synthesize sapienate impaired. Our discovery explains metabolic plasticity in fatty acid desaturation and constitutes an unexplored metabolic rewiring in cancers.- Published
- 2019
- Full Text
- View/download PDF
45. Low TREM1 expression in whole blood predicts anti-TNF response in inflammatory bowel disease.
- Author
-
Verstockt B, Verstockt S, Dehairs J, Ballet V, Blevi H, Wollants WJ, Breynaert C, Van Assche G, Vermeire S, and Ferrante M
- Subjects
- Adult, Biomarkers, Colitis, Ulcerative blood, Colitis, Ulcerative diagnosis, Colitis, Ulcerative drug therapy, Crohn Disease blood, Crohn Disease diagnosis, Crohn Disease drug therapy, Female, Gene Expression, Humans, Inflammatory Bowel Diseases diagnosis, Male, Middle Aged, Molecular Targeted Therapy, Nomograms, Prognosis, Treatment Outcome, Triggering Receptor Expressed on Myeloid Cells-1 genetics, Antibodies, Monoclonal therapeutic use, Inflammatory Bowel Diseases blood, Inflammatory Bowel Diseases drug therapy, Triggering Receptor Expressed on Myeloid Cells-1 blood, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Background: With the changed therapeutic armamentarium for Crohn's disease (CD) and ulcerative colitis (UC), biomarkers predicting treatment response are urgently needed. We studied whole blood and mucosal expression of genes previously reported to predict outcome to anti-TNF therapy, and investigated if the signature was specific for anti-TNF agents., Methods: We prospectively included 54 active IBD patients (24CD, 30UC) initiating anti-TNF therapy, as well as 22 CD patients initiating ustekinumab and 51 patients initiating vedolizumab (25CD, 26UC). Whole blood expression of OSM, TREM1, TNF and TNFR2 was measured prior to start of therapy using qPCR, and mucosal gene expression in inflamed biopsies using RNA-sequencing. Response was defined as endoscopic remission (SES-CD ≤ 2 at week 24 for CD and Mayo endoscopic sub-score ≤ 1 at week 10 for UC)., Findings: Baseline whole blood TREM1 was downregulated in future anti-TNF responders, both in UC (FC = 0.53, p = .001) and CD (FC = 0.66, p = .007), as well as in the complete cohort (FC = 0.67, p < .001). Receiver operator characteristic statistics showed an area under the curve (AUC) of 0.78 (p = .001). A similar accuracy could be achieved with mucosal TREM1 (AUC 0.77, p = .003), which outperformed the accuracy of serum TREM1 (AUC 0.58, p = .31). Although differentially expressed in tissue, OSM, TNF and TNFR2 were not differentially expressed in whole blood. The TREM1 predictive signal was anti-TNF specific, as no changes were seen in ustekinumab and vedolizumab treated patients., Interpretation: We identified low TREM-1 as a specific biomarker for anti-TNF induced endoscopic remission. These results can aid in the selection of therapy in biologic-naïve patients., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
46. Sustained SREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy.
- Author
-
Talebi A, Dehairs J, Rambow F, Rogiers A, Nittner D, Derua R, Vanderhoydonc F, Duarte JAG, Bosisio F, Van den Eynde K, Nys K, Pérez MV, Agostinis P, Waelkens E, Van den Oord J, Fendt SM, Marine JC, and Swinnen JV
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Down-Regulation, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Knockout Techniques, Humans, Lipogenesis drug effects, Male, Melanocytes, Melanoma genetics, Mice, Mice, Nude, Mice, SCID, Mutation, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf genetics, Pyridines pharmacology, Signal Transduction genetics, Sterol Regulatory Element Binding Protein 1 antagonists & inhibitors, Sterol Regulatory Element Binding Protein 1 genetics, Thiazoles pharmacology, Vemurafenib, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm genetics, Lipogenesis genetics, Melanoma drug therapy, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Sterol Regulatory Element Binding Protein 1 metabolism
- Abstract
Whereas significant anti-tumor responses are observed in most BRAF
V600E -mutant melanoma patients exposed to MAPK-targeting agents, resistance almost invariably develops. Here, we show that in therapy-responsive cells BRAF inhibition induces downregulation of the processing of Sterol Regulator Element Binding (SREBP-1) and thereby lipogenesis. Irrespective of the escape mechanism, therapy-resistant cells invariably restore this process to promote lipid saturation and protect melanoma from ROS-induced damage and lipid peroxidation. Importantly, pharmacological SREBP-1 inhibition sensitizes BRAFV600E -mutant therapy-resistant melanoma to BRAFV600E inhibitors both in vitro and in a pre-clinical PDX in vivo model. Together, these data indicate that targeting SREBP-1-induced lipogenesis may offer a new avenue to overcome acquisition of resistance to BRAF-targeted therapy. This work also provides evidence that targeting vulnerabilities downstream of oncogenic signaling offers new possibilities in overcoming resistance to targeted therapies.- Published
- 2018
- Full Text
- View/download PDF
47. CRISP-ID: decoding CRISPR mediated indels by Sanger sequencing.
- Author
-
Dehairs J, Talebi A, Cherifi Y, and Swinnen JV
- Subjects
- Algorithms, CRISPR-Cas Systems, INDEL Mutation, Internet, Gene Editing methods, Sequence Analysis, DNA methods
- Abstract
The advent of next generation gene editing technologies has revolutionized the fields of genome engineering in allowing the generation of gene knockout models and functional gene analysis. However, the screening of resultant clones remains challenging due to the simultaneous presence of different indels. Here, we present CRISP-ID, a web application which uses a unique algorithm for genotyping up to three alleles from a single Sanger sequencing trace, providing a robust and readily accessible platform to directly identify indels and significantly speed up the characterization of clones.
- Published
- 2016
- Full Text
- View/download PDF
48. Loss of Chromosome 8p Governs Tumor Progression and Drug Response by Altering Lipid Metabolism.
- Author
-
Cai Y, Crowther J, Pastor T, Abbasi Asbagh L, Baietti MF, De Troyer M, Vazquez I, Talebi A, Renzi F, Dehairs J, Swinnen JV, and Sablina AA
- Subjects
- Antineoplastic Agents pharmacology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Hypoxia, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cell Survival drug effects, Cell Survival genetics, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic genetics, Disease Progression, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, In Situ Hybridization, Fluorescence methods, Kaplan-Meier Estimate, Lipid Metabolism drug effects, Multivariate Analysis, Prognosis, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Spheroids, Cellular drug effects, Spheroids, Cellular metabolism, Breast Neoplasms genetics, Chromosome Deletion, Chromosomes, Human, Pair 8 genetics, Lipid Metabolism genetics
- Abstract
Large-scale heterozygous deletions are a hallmark of cancer genomes. The concomitant loss of multiple genes creates vulnerabilities that are impossible to reveal through the study of individual genes. To delineate the functional outcome of chromosome 8p loss of heterozygosity (LOH), a common aberration in breast cancer, we modeled 8p LOH using TALEN-based genomic engineering. 8p LOH alters fatty acid and ceramide metabolism. The shift in lipid metabolism triggers invasiveness and confers tumor growth under stress conditions due to increased autophagy. The resistance of 8p-deleted cells to chemotherapeutic drugs concurs with poorer survival rates of breast cancer patients harboring an 8p LOH. The autophagy dependency of 8p-deleted cells provides the rational basis for treatment of 8p LOH tumors with autophagy inhibitors., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. Phospholipid profiling identifies acyl chain elongation as a ubiquitous trait and potential target for the treatment of lung squamous cell carcinoma.
- Author
-
Marien E, Meister M, Muley T, Gomez Del Pulgar T, Derua R, Spraggins JM, Van de Plas R, Vanderhoydonc F, Machiels J, Binda MM, Dehairs J, Willette-Brown J, Hu Y, Dienemann H, Thomas M, Schnabel PA, Caprioli RM, Lacal JC, Waelkens E, and Swinnen JV
- Subjects
- Animals, Fatty Acid Elongases, Heterografts, Humans, Mice, Acetyltransferases metabolism, Carcinoma, Squamous Cell chemistry, Lung Neoplasms chemistry, Phospholipids chemistry
- Abstract
Lung cancer is the leading cause of cancer death. Beyond first line treatment, few therapeutic options are available, particularly for squamous cell carcinoma (SCC). Here, we have explored the phospholipidomes of 30 human SCCs and found that they almost invariably (in 96.7% of cases) contain phospholipids with longer acyl chains compared to matched normal tissues. This trait was confirmed using in situ 2D-imaging MS on tissue sections and by phospholipidomics of tumor and normal lung tissue of the L-IkkαKA/KA mouse model of lung SCC. In both human and mouse, the increase in acyl chain length in cancer tissue was accompanied by significant changes in the expression of acyl chain elongases (ELOVLs). Functional screening of differentially expressed ELOVLs by selective gene knockdown in SCC cell lines followed by phospholipidomics revealed ELOVL6 as the main elongation enzyme responsible for acyl chain elongation in cancer cells. Interestingly, inhibition of ELOVL6 drastically reduced colony formation of multiple SCC cell lines in vitro and significantly attenuated their growth as xenografts in vivo in mouse models. These findings identify acyl chain elongation as one of the most common traits of lung SCC discovered so far and pinpoint ELOVL6 as a novel potential target for cancer intervention.
- Published
- 2016
- Full Text
- View/download PDF
50. Identification of drugs that restore primary cilium expression in cancer cells.
- Author
-
Khan NA, Willemarck N, Talebi A, Marchand A, Binda MM, Dehairs J, Rueda-Rincon N, Daniels VW, Bagadi M, Thimiri Govinda Raj DB, Vanderhoydonc F, Munck S, Chaltin P, and Swinnen JV
- Subjects
- A549 Cells, Antineoplastic Agents classification, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Cycle drug effects, Cell Line, Tumor, Cilia metabolism, Gefitinib, Humans, Microscopy, Confocal, Neoplasms metabolism, Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Quinazolines pharmacology, Reproducibility of Results, Antineoplastic Agents pharmacology, Cell Proliferation drug effects, Cilia drug effects, Drug Screening Assays, Antitumor methods
- Abstract
The development of cancer is often accompanied by a loss of the primary cilium, a microtubule-based cellular protrusion that functions as a cellular antenna and that puts a break on cell proliferation. Hence, restoration of the primary cilium in cancer cells may represent a novel promising approach to attenuate tumor growth. Using a high content analysis-based approach we screened a library of clinically evaluated compounds and marketed drugs for their ability to restore primary cilium expression in pancreatic ductal cancer cells. A diverse set of 118 compounds stimulating cilium expression was identified. These included glucocorticoids, fibrates and other nuclear receptor modulators, neurotransmitter regulators, ion channel modulators, tyrosine kinase inhibitors, DNA gyrase/topoisomerase inhibitors, antibacterial compounds, protein inhibitors, microtubule modulators, and COX inhibitors. Certain compounds also dramatically affected the length of the cilium. For a selection of compounds (Clofibrate, Gefitinib, Sirolimus, Imexon and Dexamethasone) their ability to restore ciliogenesis was confirmed in a panel of human cancer cell line models representing different cancer types (pancreas, lung, kidney, breast). Most compounds attenuated cell proliferation, at least in part through induction of the primary cilium, as demonstrated by cilium removal using chloral hydrate. These findings reveal that several commonly used drugs restore ciliogenesis in cancer cells, and warrant further investigation of their antineoplastic properties.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.