nutrients

Article FABP7 Facilitates Uptake of Docosahexaenoic Acid in Glioblastoma Neural Stem-like Cells

Won-Shik Choi 1,†, Xia Xu 1,†, Susan Goruk 2, Yixiong Wang 1, Samir Patel 1, Michael Chow 3, Catherine J. Field 2 and Roseline Godbout 1,*

1 Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada; [email protected] (W.-S.C.); [email protected] (X.X.); [email protected] (Y.W.); [email protected] (S.P.) 2 Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; [email protected] (S.G.); cjfi[email protected] (C.J.F.) 3 Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada; [email protected] * Correspondence: [email protected]; Tel.: +780-432-8901; Fax: +780-432-8892 † These authors contribute equally to this work.

Abstract: Glioblastoma (GBM) is an aggressive tumor with a dismal prognosis. Neural stem-like cells contribute to GBM’s poor prognosis by driving drug resistance and maintaining cellular heterogene- ity. GBM neural stem-like cells express high levels of brain fatty acid-binding (FABP7), which binds to polyunsaturated fatty acids (PUFAs) ω-6 arachidonic acid (AA) and ω-3 docosahexaenoic acid (DHA). Similar to brain, GBM tissue is enriched in AA and DHA. However, DHA levels are considerably lower in GBM tissue compared to adult brain. Therefore, it is possible that increasing DHA content in GBM, particularly in neural stem-like cells, might have therapeutic value. Here, we examine the fatty acid composition of patient-derived GBM neural stem-like cells grown as neuro-

 sphere cultures. We also investigate the effect of AA and DHA treatment on the fatty acid profiles of  GBM neural stem-like cells with or without FABP7 knockdown. We show that DHA treatment in-

Citation: Choi, W.-S.; Xu, X.; Goruk, creases DHA levels and the DHA:AA ratio in GBM neural stem-like cells, with FABP7 facilitating the S.; Wang, Y.; Patel, S.; Chow, M.; Field, DHA uptake. We also found that an increased uptake of DHA inhibits the migration of GBM neural C.J.; Godbout, R. FABP7 Facilitates stem-like cells. Our results suggest that increasing DHA content in the GBM microenvironment may Uptake of Docosahexaenoic Acid in reduce the migration/infiltration of FABP7-expressing neural stem-like cancer cells. Glioblastoma Neural Stem-like Cells. Nutrients 2021, 13, 2664. https:// Keywords: glioblastoma; docosahexaenoic acid; neural stem-like cells; B-FABP; FABP7; fatty acids; doi.org/10.3390/nu13082664 phospholipids; neurospheres

Academic Editor: Asim K. Duttaroy

Received: 30 June 2021 1. Introduction Accepted: 29 July 2021 Published: 30 July 2021 Glioblastoma (grade IV astrocytoma) is the most common primary brain cancer in adults [1,2]. It is a highly aggressive and deadly cancer, with a median survival time of

Publisher’s Note: MDPI stays neutral ~15 months despite multimodality therapy [3,4]. Resistance to treatment is due in part to with regard to jurisdictional claims in the invasion of surrounding brain parenchyma by GBM cells, suggesting that targeting the published maps and institutional affil- infiltrative properties of GBM cells might be an effective therapeutic strategy [5]. In addition iations. to their infiltrative nature, GBMs are highly heterogeneous tumors at both the cellular and molecular levels, adding an additional level of complexity to the effective targeting of these tumors [6,7]. Multi-lineage differentiation from GBM neural stem-like cells forms the basis of GBM heterogeneity [8,9], with GBM neural stem-like cells displaying resistance to radiation and chemotherapy, thereby repopulating the tumor [10,11]. GBM neural stem- Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland. like cells exhibit distinct metabolic profiles compared to non-stem cell populations, with This article is an open access article reduced glycolysis and higher lipid metabolism [12–14]. Dysregulation of lipid metabolism distributed under the terms and has also been associated with maintenance of GBM stemness and poor survival [14,15]. conditions of the Creative Commons Important mediators of lipid metabolism, such as brain fatty acid-binding protein (B-FABP Attribution (CC BY) license (https:// or FABP7) [16], fatty acid transporter CD36 [17], acyl-CoA-binding protein (ACBP) [18], creativecommons.org/licenses/by/ and lipid elongation enzyme ELOVL2 [19] have all been reported to be highly expressed in 4.0/). GBM neural stem-like cells, highlighting the importance of lipid metabolism.

Nutrients 2021, 13, 2664. https://doi.org/10.3390/nu13082664 https://www.mdpi.com/journal/nutrients Nutrients 2021, 13, 2664 2 of 16

The human adult brain is highly enriched in lipids, especially long chain polyunsat- urated fatty acids (PUFAs) [20,21]. The two main types of PUFAs in the brain are ω-6 arachidonic acid (AA) and ω-3 docosahexaenoic acid (DHA) [20]. DHA and AA are be- lieved to have opposing roles in cancer [22]. AA is the precursor of ω-6 series eicosanoids such as prostaglandin E2 (PGE2), which stimulate inflammation, cancer growth, and in- vasion [23–25]. In contrast, DHA is the precursor of neuroprotectins and resolvins, which resolve inflammation and inhibit cancer growth [26–28]. In healthy adult brain, there is a tightly controlled balance between ω-6 and ω-3 PUFAs, with DHA levels exceeding AA levels in both total lipids and total phospholipids [29]. In human GBM tumor tissue, AA levels remain similar to that of healthy brain tissue; however, DHA levels are decreased by 50%, resulting in a significantly lower DHA:AA ratio [30,31]. DHA supplementation in GBM cells inhibits cell proliferation [32,33] and migration [34,35], and induces apopto- sis [32,35,36]. Furthermore, DHA sensitizes GBM cells to ionizing radiation [37], suggesting that DHA supplementation may benefit GBM patients. Brain fatty acid-binding protein (B-FABP or FABP7) is normally expressed in neural stem cells during development [38,39], and is also expressed in GBM stem-like cells [16,40]. FABP7, whose preferred ligands are PUFAs [41], is preferentially found at the infiltrative edges of GBM tumors [16,42] and its expression correlates with increased GBM cell mi- gration [43]. DHA treatment has previously been shown to increase DHA content in the total lipids of U87 GBM cells [32]. Furthermore, C6 glioma cells injected directly into rat brain have elevated ω-3 PUFA levels and an increased ω-3:ω-6 ratio when rats are fed a DHA-rich diet compared to control diet [44]. Based on our previous work, DHA inhibits GBM cell migration in a FABP7-dependent manner [34]. However, these results were obtained with well-established cell lines believed to represent the more ‘differentiated’ aspect of GBM tumors. As resistance to treatment and poor prognosis has been linked to the neural stem-like tumor cell populations (a.k.a. brain tumor-initiating cells) in GBM tumors [10,11,45,46], it is important to know whether DHA treatment will have similar effects on GBM neural stem-like cells. Here, we examine the fatty acid composition of patient-derived GBM cells cultured in either regular medium (contains fetal calf serum and promotes a generally more differenti- ated phenotype) or neurosphere medium (contains B-27 supplement and growth factors and promotes a more neural stem cell-like phenotype) [47,48]. We compare the effects of DHA treatment and FABP7 expression on DHA uptake and cell migration in these two types of GBM cells. We also investigate the role of FABP7, which is highly expressed in GBM stem-like cells [40], in the uptake of DHA in lipids and phospholipids. Our results indicate that DHA treatment increases the DHA content in total lipids and phospholipids of GBM neural stem-like cells, with FABP7 expression facilitating DHA uptake. We also report a correlation between increased DHA uptake and decreased cell migration in GBM neural stem-like cells.

2. Materials and Methods 2.1. Primary GBM Neurosphere Cultures GBM primary cultures (A4-004, A4-007, A4-011, and A4-012) were prepared by en- zymatic digestion of GBM biopsies obtained from patients who were consented prior to surgery under Health Research Ethics Board of Alberta Cancer Committee approval (HREBA.CC-14-0070). GBM cells were plated in either DMEM supplemented with 10% fetal calf serum (FCS) (for the generation of adherent cultures with a more differentiated phenotype) or DMEM/F12 medium supplemented with 0.5X B-27 (Life Technologies, Carlsbad, CA, USA), 20 ng/mL epidermal growth factor (EGF), and 10 ng/mL fibroblast growth factor (FGF) (for the generation of neurosphere cultures with a neural stem cell- like phenotype). ED501 neurosphere cultures and U251 GBM cells have been previously described [49]. Nutrients 2021, 13, 2664 3 of 16

2.2. Establishment of Stable FABP7-Depleted Cell Lines Lentivirus shRNA packaging plasmids and control plasmids were purchased from Sigma. The two lentivirus FABP7 shRNA constructs used for our experiments were obtained from the University of Alberta RNAi Core Facility. FABP7 shRNA constructs sequences and virus production have been described previously [49]. The MISSION pLKO.1 plasmid (SHC002; Sigma-Aldrich, St. Louis, MO, USA) served as the control vector. A4-004N, U251 and ED501 cells were infected with lentivirus overnight and the medium changed. Infected GBM cells were selected in 1 µg/mL puromycin.

2.3. Western Blotting For western blot analysis, we loaded 40 µg of whole cell lysates per lane. from whole cell lysates were separated by SDS-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes. Membranes were then immunoblotted with rabbit polyclonal anti-FABP7 (prepared in-house; 1:1000) [49] and mouse anti-GAPDH (1:1000; Thermo Fisher Scientific, Waltham, MA, USA) antibodies, followed by horseradish peroxidase-conjugated secondary antibodies (1:50,000; Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA) using ECL Western Detection Reagent (GE Healthcare Life Sciences, Chicago, IL, USA).

2.4. Fatty Acid Preparation and Treatment Fatty acids (DHA and AA) (Sigma-Aldrich, St. Louis, MO, USA) were dissolved in ethanol, then complexed to BSA (Roche) over a steady stream of nitrogen gas and stored at −80 ◦C under reducing conditions. Both GBM neurosphere and GBM adherent cells ◦ were cultured at 37 C in a humidified 5% CO2 atmosphere. For fatty acid treatment, A4-004 neurosphere and adherent cells at 60–70% confluency were cultured in medium (neurosphere medium for A4-004N and serum-free DMEM for A4-004Adh) with 30 µM BSA (vehicle control), 30 µM DHA, or 30 µM AA for 24 h. Cells were then collected for lipid extraction and fatty acid analysis. Experiments were repeated three times.

2.5. Lipid Extraction and Gas Chromatography Total lipids were extracted with chloroform/methanol (2:1 vol/vol) using a modifica- tion of the Folch procedure [50]. Thin layer chromatography on silica G plates was used to isolate total phospholipids. Samples were scraped and methylated at 110 ◦C with a mixture (1:1) of BF3/methanol reagent (Sigma) and hexane. Fatty acid methyl esters were separated and identified by gas liquid chromatography (Agilent Model7890A, Agilent Technologies) using a 100 m CP-Sil 88 fused capillary column (100 m × 0.25 mm, Agilent Technologies, Santa Clara, CA, USA) and STD 502 (NuChek, Elysian, MN, USA) [51]. Fatty acids with a 14 to 24 carbon chain length were quantified and are presented as % of total fatty acids.

2.6. Semi-Quantitative RT-PCR RNA was purified from paired patient-derived GBM neurosphere (A4-004N, A4- 007N, A4-011N and A4-012N) and adherent cells (A4-004Adh, A4-007Adh, A4-011Adh and A4-012Adh) using the RNeasy Plus Kit (Qiagen, Hilden, Germany), and cDNA was generated using Superscript II reverse transcriptase (Life Technologies). The following primers were used for semi-quantitative RT-PCR analysis: FABP7 (Forward 50-TGGAGGCTTTCTGTGCTAC-30; Reverse 50-TAGGATAGCACTGAGACTTG-30), SOX- 2 (Forward 50-ACACTGCCCCTCTCACACA-30; Reverse 50-CATTTTTTTCGTCGCTTGGAG- 30), Nestin (Forward 50-GGAGAAACAGGGCCTACAG-30; Reverse 50-GCAGAGAGAGA GGAGCATC-30) and β-actin (Forward 50-CTGGCACCACACCTTCTAC-30; Reverse 50- CATACTCCTGCTTGCTGATC-30).

2.7. Lipid Droplet Analysis GBM neural stem-like cells (A4-004N and ED501) were cultured on coverslips in neurosphere medium followed by treatment with BSA (vehicle control) or 30 µM DHA for Nutrients 2021, 13, 2664 4 of 16

24 h. U251 cells were cultured in serum-free DMEM supplemented with BSA or 60 µM DHA for 24 h. Cells were fixed with 4% paraformaldehyde for 5 min at room temperature and stained with 1 µg/mL Nile Red for 15 min. Coverslips were mounted with Mowiol 4-88 mounting medium containing DAPI and images were acquired using a Zeiss confocal microscope. ImageJ software was used for quantitative analysis. We used particle analysis to identify the number of nuclei (indicative of number of cells) in each image. The images were thresholded so that particles >20 µm2 were identified as nuclei. The Nile Red channel was thresholded to minimize background signal and identify regions of interest (ROIs). Nile Red total intensity was calculated based on the number of pixels under each intensity (0–255) in the ROIs of each image. The average Nile Red intensity per cell was calculated based on total Nile Red intensity divided by the number of cells in each image.

2.8. Transwell Assay Directional cell migration was measured using the Transwell cell migration assay. Fifty thousand cells in neurosphere medium were seeded in the top chambers of 24-well cell culture Transwell inserts (Falcon Cell Culture Inserts, Corning, Glendale, AZ, USA). Cells were allowed to migrate through an 8-µm polyethylene terephthalate (PET) membrane towards a chemoattractant (DMEM + 10% fetal calf serum) in the bottom chamber for 20 h. Fatty acids were added to culture medium for 24 h before carrying out the migration assay. Cell fixation, immunostaining and cell counts have been previously described [52]. Three independent experiments were carried out for each cell line tested.

2.9. Statistical Analysis Assessment of the significance of differences between groups was by one-way ANOVA followed by post-hoc Tukey’s test (three groups comparison) and student’s unpaired t-test (two groups comparison). Microsoft Excel (Microsoft, Redmond, WA, USA) and Prism 8 (GraphPad Software, San Diego, CA, USA) were used for statistical analysis of data. A p-value < 0.05 was considered statistically significant.

3. Results 3.1. Fatty Acid Profiles of Total Lipids Extracted from GBM Neural Stem-like versus Adherent Cells GBM tumor tissues have elevated levels of proteins involved in fatty acid metabolism compared to normal brain [53]. Many of these proteins (FABP7, FASN and ELOVL2) are up- regulated in GBM patient-derived tumor stem cell cultures compared to more-differentiated adherent cells cultured in serum-containing medium [13,19,40]. Expression of PUFA metabolism in GBM neural stem-like cells suggests that it may be possible to alter the PUFA content of these cells through manipulation of the lipid microenvironment. As our previous results indicate that an increased DHA:AA ratio inhibits GBM cell migration [34], we compared the fatty acid profiles of GBM neural stem-like cells versus GBM adherent cells, and the effect of DHA and AA supplementation on their respective fatty acid profiles. We established GBM cultures from patients using “neurosphere” medium (promotes growth of neural stem-like cells) and “FCS-containing” medium (promotes growth of GBM cells with a more differentiated adherent phenotype) [48]. We first examined the expression of FABP7, an established GBM neural stem-like cell marker [40], in our paired GBM cultures using semi-quantitative RT-PCR. While all eight cultures expressed FABP7, levels were generally higher in the neural stem-like cell cultures compared to adherent cultures (Figure1). Similar trends were observed when we examined the expression of the well characterized GBM neural stem cell markers, Nestin and SOX-2 (Figure1). These results are consistent with a previous report showing higher expression of FABP7 in neurosphere cultures compared to adherent cultures, and correlation of FABP7 with Nestin and SOX-2 [40]. As the biggest difference in FABP7 levels was observed in A4-004, we used A4-004N and A4-004Adh cultures for our fatty acid profiling study. Nutrients 2021, 13, 2664 5 of 15

expression of FABP7, an established GBM neural stem-like cell marker [40], in our paired GBM cultures using semi-quantitative RT-PCR. While all eight cultures expressed FABP7, levels were generally higher in the neural stem-like cell cultures compared to adherent cultures (Figure 1). Similar trends were observed when we examined the expression of the well characterized GBM neural stem cell markers, Nestin and SOX-2 (Figure 1). These re- sults are consistent with a previous report showing higher expression of FABP7 in neuro- Nutrients 2021, 13, 2664 sphere cultures compared to adherent cultures, and correlation of FABP7 with Nestin5 and of 16 SOX-2 [40]. As the biggest difference in FABP7 levels was observed in A4-004, we used A4-004N and A4-004Adh cultures for our fatty acid profiling study.

Figure 1. Preferential expression of FABP7 in neurosphere cultures. Semi-quantitative RT-PCR Figure 1. Preferential expression of FABP7 in neurosphere cultures. Semi-quantitative RT-PCR anal- analysis of FABP7, SOX-2, and Nestin (NES) in paired patient-derived GBM adherent and neurosphere ysis of FABP7, SOX-2, and Nestin (NES) in paired patient-derived GBM adherent and neurosphere cultures. β-Actin was used as the loading control. cultures. β-Actin was used as the loading control. Total lipids were extracted from A4-004N and A4-004Adh cells cultured in their re- spectiveTotal media lipids and were fatty extracted acid composition from A4-004N was analysed and A4-004Adh by gas chromatography cells cultured in (Table their S1).re- spectiveDifferences media in fatty and acidfatty composition acid composition were observed was analysed for all by classes gas chromatography of fatty acids (saturated (Table S1).fatty Differences acids (SFAs), in fatty monounsaturated acid composition fatty were acids observed (MUFAs), for and all polyunsaturatedclasses of fatty acidsω-6 (sat- and uratedω-3 PUFAs)) fatty acids (Table (SFAs), S1 and monounsaturated Figure2A). The mostfatty prevalentacids (MUFAs),ω-3 PUFA and polyunsaturated in A4-004Adh was ω- 6DHA and ω (1%-3 PUFAs)) of total fatty (Table acids), S1 and while Figure DHA 2A). was The virtually most prevalent undetectable ω-3 PUFA in A4-004N in A4-004Adh (Figure was2C). DHA In A4-004N, (1% of thetotal most fatty abundant acids), whileω-3 PUFADHA was was virtually C18:3 (alpha-linolenic undetectable acidin A4-004N (ALA)) (Figure(1.79% of2C). total In fatty A4-004N, acids) (Tablethe most S1). abundant The most prevalentω-3 PUFA forms was ofC18:3ω-6 (alpha-linolenic PUFA were C20:2 acidω-6 (ALA))(3.25% of(1.79% total of fatty total acids fatty in acids) A4-004N (Table vs. S1). 0.95% The in most A4-004Adh) prevalent and forms C20:4 of ωω-6-6 (AA)PUFA (1.01% were C20:2in A4-004Nω-6 (3.25% vs. 3.01% of total in A4-004Adh)fatty acids in (Figure A4-004N2D). vs. The 0.95% DHA:AA in A4-004Adh) ratio was much and C20:4 lowerω in-6 (AA)A4-004N (1.01% compared in A4-004N to A4-004Adh vs. 3.01% cellsin A4-004Adh) (0.14 vs. 0.84), (Figure whereas 2D). the Theω -3:DHA:AAω-6 ratio ratio showed was muchno difference lower in in A4-004N A4-004N compared compared to to A4-004Adh A4-004Adh cells cells (0.14 (0.42 vs. vs. 0.84), 0.41) whereas (Figure2 E).the ω-3:ω- 6 ratioNext, showed we examinedno difference the effectin A4-004N of AA comp or DHAared treatment to A4-004Adh on the cells fatty (0.42 acid vs. composition 0.41) (Fig- ureof A4-004N 2E). and A4-004Adh total lipids (Table S2). Cells were cultured in their respective “neurosphere”Next, we examined versus “adherent” the effect of media, AA or supplemented DHA treatment with on the BSA fatty (control), acid composition 30 µM AA ofor A4-004N 30 µM DHA and A4-004Adh for 24 h before total they lipids were (Table harvested S2). Cells for were lipid cultured extraction. in their To ensure respective that “neurosphere”BSA itself would versus not affect“adherent” our results, media, we supplemented also compared with the BSA fatty (control), acid composition 30 μM AA or of 30A4-004N μM DHA and for A4-004Adh 24 h before lipids they in were BSA harvested (vehicle control) for lipid and extraction. untreated To cells. ensure No significant that BSA itselfdifferences would were not affect noted our between results, untreated we also and compared BSA-treated the fatty cells acid (Table composition S3). of A4- 004NAA and treatment A4-004Adh increased lipids in AA BSA levels, (vehicle along control) with itsand downstream untreated cells. mediators, No significant adrenic differencesacid (ADA, were C22:4 notedω-6), between and docosapentaenoic untreated and acid BSA-treated (DPA, C22:5 cellsω (Table-6) in both S3). A4-004N and A4-004AdhAA treatment cells (Table increased S2). TheAA DHA:AAlevels, along ratio wi wasth its decreased downstream ~7-fold mediators, and 3-fold adrenic upon acidAA treatment(ADA, C22:4 in A4-004Nω-6), and and docosapentaenoic A4-004Adh, respectively acid (DPA, (Figure C22:5ω3C),-6) whereasin both A4-004N the ω-3: andω-6 A4-004Adhratio was decreased cells (Table ~3-fold S2). The and 2-foldDHA:AA upon rati AAo was treatment decreased in A4-004N ~7-fold and and 3-fold A4-004Adh, upon AArespectively treatment (Figure in A4-004N3D). and A4-004Adh, respectively (Figure 3C), whereas the ω-3:ω-6 ratio Strikingly,was decreased DHA ~3-fold treatment and increased 2-fold upon DHA AA content treatment by more in A4-004N than 100-fold and A4-004Adh, (an increase respectivelyfrom 0.1% of (Figure fatty acids 3D). to 11.2% of fatty acids) in A4-004N total lipids compared to 8-fold (1.01% to 8.09%) in A4-004Adh (Figure3A). This increase in DHA resulted in an ~130-fold increase in the DHA:AA ratio in A4-004N (from 0.07 to 9.65) compared to ~9-fold in A4- 004Adh cells (Figure3C). The overall increase in the ω-3:ω-6 ratio was ~6-fold and ~4-fold in A4-004N and A4-004Adh DHA-treated cells, respectively (Figure3D). These results indicate efficient uptake of both AA and DHA in neural stem cell-like cultures.

Nutrients 2021, 13, 2664 6 of 15

Nutrients 2021, 13, 2664 6 of 15 Nutrients 2021, 13, 2664 6 of 16

Figure 2. Fatty acid composition of total lipids and total phospholipids extracted from A4-004N and A4-004Adh cells. Total lipidsFigure (TL) 2.wereFatty extracted, acid composition and total of total phospholipids lipids and total (PL) phospholipids separated extracted from A4-004N from A4-004N and and A4-004Adh A4-004Adh and cells. fatty acid compositionFigure 2. TotalFatty measured lipids acid (TL) composition by were gas extracted, chromatography. of total and totallipids phospholipids (andA) Percent total phosph (PL) abundance separatedolipids of from extractedsaturated A4-004N from fa andtty A4-004Adh A4-004Nacids (SFA), andand monounsaturated fatty A4-004Adh acid cells. fattyTotal acids lipids composition(MUFA), (TL) were and measured extracted, polyunsa by gas turatedand chromatography. total fatty phospholipids acids (A) Percent (PUFA) abundance(PL) in totalseparated of lipids saturated from and fatty totalA4-004N acids phos (SFA), pholipidsand monounsaturated A4-004Adh of A4-004N and fatty fatty and A4-acid 004Adhcomposition cells.acids measured(B (MUFA),) Percent and by abundance polyunsaturated gas chromatography. of ω fatty-3 PUFA acids ( (PUFA)Aand) Percent ω in-6 totalfatty abundance lipids acids and PUFA total of phospholipidssaturated in total lipids fatty of A4-004Nand acids total (SFA), and phospholipids A4-004Adh monounsaturated of A4- ω ω 004Nfatty andacids A4-004Adhcells. (MUFA), (B) Percent and cells. abundancepolyunsa (C) Percent ofturated-3 abundance PUFA fatty and acids of-6 fattyC20:5(PUFA) acidsω-3 in PUFA(EPA) total in lipidsand total C22:6 lipids andω andtotal-3 total(DHA) phos phospholipidspholipids fatty acids of in A4-004NA4-004N total lipids and and A4- and A4-004Adh cells. (C) Percent abundance of C20:5ω-3 (EPA) and C22:6ω-3 (DHA) fatty acids in total lipids and total total004Adh phospholipids cells. (B) Percent of A4-004N abundance and A4-004Adh of ω-3 PUFA cells. and ( ωD-6) Percent fatty acids abundance PUFA in of total C20:4 lipidsω-6 and(AA), total C20:2 phospholipidsω-6 and C20:3 ofω A4--6 phospholipids of A4-004N and A4-004Adh cells. (D) Percent abundance of C20:4ω-6 (AA), C20:2ω-6 and C20:3ω-6 (DGLA) (DGLA)004N and fatty A4-004Adh acids in total cells. lipids (C) Percent and total abundance phospholipids of C20:5 of A4-004Nω-3 (EPA) and and A4-004Adh C22:6ω-3 (DHA)cells. (E fatty) DHA:AA acids inratio, total and lipids ω-3: andω- 6 ratios in fattytotal acids lipids in totaland lipids total and phospholipids total phospholipids of A4-004N of A4-004N an andd A4-004Adh A4-004Adh cells. cells. (E) See DHA:AA Tables ratio, S1 and andω -3:S4ω for-6 ratios comprehensive in total phospholipidstotal lipids and of totalA4-004N phospholipids and A4-004Adh of A4-004N cells. and A4-004Adh (D) Percent cells. abundance See Tables S1 of and C20:4 S4 forω comprehensive-6 (AA), C20:2 listsω of-6 fattyand C20:3ω-6 lists(DGLA) of fatty fatty acids acids in intotal total lipids lipids and and phospholipids, total phospholipids respectively. of A4-004N n = 3. *and indicates A4-004Adh p < 0.05, cells. ** indicates (E) DHA:AA p < 0.01, ratio, *** and indicates ω-3:ω- p < 0.001, andacids **** in totalindicates lipids and p < phospholipids,0.0001. Abbreviations: respectively. DHA,n = 3. *docosahe indicates pxaenoic< 0.05, ** acid; indicates AA,p

Figure 3. Effects of PUFA treatment on AA- and DHA-incorporation in total lipids and total phospholipids from A4-004N Figure 3. Effectsand A4-004Adh of PUFA cells. treatment A4-004N on and AA- A4-004Adh and DHA-incorporatio culture media were supplementedn in total lipids with and BSA (control),total phospholipids 30 µM AA, or 30fromµM A4-004N and A4-004AdhDHA. Percentcells. A4-004N abundance and of DHA A4-004Adh (A,E), percent cultur abundancee media of were AA ( Bsupplemented,F), DHA:AA ratio with (C,G BSA) and (control),ω-3:ω-6 ratio 30 (μDM,H )AA, or 30 in total lipids (A–D) and total phospholipids (E–H) of A4-004N and A4-004Adh cells. n = 3. Different letters indicate μFigureM DHA. 3. EffectsPercent of abundance PUFA treatment of DHA on (A AA-,E), percentand DHA-incorporatio abundance of AAn in (B total,F), DHA:AAlipids and ratio total ( Cphospholipids,G) and ω-3:ω from-6 ratio A4-004N (D,H) in total lipidsthat ( groupsA–D) and are significantly total phospholipids different. See (E– TablesH) of S2A4-004N and S5 for and comprehensive A4-004Adh lists cells. of fattyn = 3. acids Different in total letters lipids andindicate that and A4-004Adhphospholipids, cells. A4-004N respectively. and A4-004Adh culture media were supplemented with BSA (control), 30 μM AA, or 30 groupsμM DHA. are Percentsignificantly abundance different. of DHA See Tables (A,E), S2percent and S5 abundance for comprehensive of AA (B,F lists), DHA:AA of fatty ratioacids ( Cin, Gtotal) and lipids ω-3: ωand-6 ratio phospho- (D,H) lipids,in total respectively. lipids (A–D ) and total phospholipids (E–H) of A4-004N and A4-004Adh cells. n = 3. Different letters indicate that groups are significantly different. See Tables S2 and S5 for comprehensive lists of fatty acids in total lipids and phospho- lipids, respectively. Strikingly, DHA treatment increased DHA content by more than 100-fold (an in- crease from 0.1% of fatty acids to 11.2% of fatty acids) in A4-004N total lipids compared to 8-foldStrikingly, (1.01% toDHA 8.09%) treatment in A4-004Adh increased (Figur DHAe 3A). content This increaseby more in than DHA 100-fold resulted (an in anin- ~130-foldcrease from increase 0.1% ofin fattythe DHA:AA acids to 11.2% ratio inof A4-004Nfatty acids) (from in A4-004N 0.07 to 9.65) total compared lipids compared to ~9- foldto 8-fold in A4-004Adh (1.01% to cells8.09%) (Figure in A4-004Adh 3C). The overall(Figure increase3A). This in increase the ω-3: inω -6DHA ratio resulted was ~6-fold in an and~130-fold ~4-fold increase in A4-004N in the andDHA:AA A4-004Adh ratio in DHA-tr A4-004Neated (from cells, 0.07 respectively to 9.65) compared (Figure to3D). ~9- fold in A4-004Adh cells (Figure 3C). The overall increase in the ω-3:ω-6 ratio was ~6-fold and ~4-fold in A4-004N and A4-004Adh DHA-treated cells, respectively (Figure 3D).

Nutrients 2021, 13, 2664 7 of 16

3.2. Fatty Acid Profiles of Phospholipids from GBM Neural Stem-like versus Adherent Cells Phospholipids are a major component of cell membranes. PUFA supplementation affects membrane phospholipid composition and fatty acid recycling [54,55]. In turn, PUFAs in cell membranes affect their properties, including fluidity and distribution of membrane-bound proteins [49,56–58]. We therefore examined the fatty acid composition of phospholipids from A4-004N and A4-004Adh cells (Table S4 and Figure2). Trends similar to those observed in total lipids were seen in phospholipids, such as increased MUFAs (42.8% vs. 39.2%) and decreased PUFAs (9.9% vs. 14.4%) in A4-004N compared to A4-004Adh cells (Table S4 and Figure2A). There was less ω-6 PUFA incorporated into the phospholipids from A4-004N (7.1%) compared to A4-004Adh cells (9.9%). Similarly, less ω-3 PUFAs were incorporated in the phospholipids from A4-004N (2.9%) compared to A4-004Adh cells (4.6%) (Figure2B). Levels of ω-3 PUFAs were similar in total lipids and phospholipids of both A4-004N and A4-004Adh cells, whereas ω-6 PUFAs were more abundant in phospholipids compared to total lipids in A4-004Adh cells, suggesting preferential incorporation of ω-6 PUFAs in membrane phospholipids of adherent cultures (Figure2B). Similar to that observed for total lipids, AA was the most abundant ω-6 PUFA in A4-004Adh phospholipids (3.8%), whereas C20:2ω-6 was the most abundant ω-6 PUFA in A4-004N total phospholipids (3.9%) (Table S4, Figure2D). Similar to total lipids, ALA (1.6%) was also abundant in A4-004N phospholipids (Table S4). Increased levels of DHA in both A4-004N and A4-004Adh total phospholipids (0.2% and 1.8%, respectively) compared to total lipids (undetectable and 1%, respectively) were noted, suggesting preferential incor- poration of DHA in phospholipids when cells are grown in their standard culture media (Figure2C). Similar to total lipids, the DHA:AA ratio was lower in A4-004N compared to A4-004Adh phospholipids (0.2 vs. 0.5). The overall ω-3:ω-6 ratio in A4-004N phospholipids was similar to that observed in A4-004Adh (0.4 vs.0.5) (Table S4 and Figure2E). AA treatment resulted in ~6X and ~2X increases in the AA content of phospholipids extracted from A4-004N and A4-004Adh cells, respectively, whereas DHA treatment in- creased the DHA content of phospholipids by ~18X and ~3.8X in A4-004N and A4-004Adh cells, respectively (Table S5). Similar to what was observed in total lipids, AA treatment resulted in ~14-fold and ~5-fold increases in the levels of the AA downstream product, ADA, in A4-004N and A4-004Adh phospholipids, respectively. In DHA-treated cells, the DHA:AA ratio increased by ~21-fold in A4-004N phospholipids, resulting in DHA:AA ratio of 4.9 (Figure3G). In comparison, there was a ~3.8-fold increase in the ratio of DHA:AA in the phospholipids of DHA-treated A4-004Adh cells compared to BSA control cells, resulting in a DHA:AA ratio of 1.8 (Figure3G). The overall increase in the phospholipid ω-3:ω-6 ratio was ~3-fold and ~2-fold in A4-004N and A4-004Adh DHA-treated cells, respectively (Figure3H). Thus, while highly significant, the incorporation of DHA into A4-004N and A4-004Adh phospholipids was not as strikingly different as that observed for total lipids, suggesting that DHA levels may be relatively more stable in phospholipids compared to total lipids in GBM cells.

3.3. FABP7 Facilitates DHA Uptake in GBM Neural Stem-like Cells FABPs play an important role in lipid-mediated biological processes through the regulation of fatty acid uptake, storage, and trafficking fatty acids to different locations in the cell [59,60]. PUFAs, especially DHA and AA, are preferred ligands for FABP7, with ω-3 DHA having a 4-fold higher binding affinity for FABP7 compared to ω-6 AA based on Isothermal Titration Calorimetry and Lipidex 1000 [41,61]. To gain insight into the role of FABP7 in the uptake of ω-3 DHA or ω-6 AA in GBM neural stem-like cells, we investigated the effect of FABP7 knockdown on DHA and AA incorporation in the total lipids and phospholipids of GBM neural stem-like cells. FABP7 was knocked-down in A4-004N cells using lentiviral shRNA constructs (shFABP7-1 and shFABP7-2) (Figure4). In A4-004N shControl cells, AA and DHA treatment increased the intracellular levels of AA and DHA, respectively, in both total lipid and total phospholipid Nutrients 2021, 13, 2664 8 of 15

Nutrients 2021, 13, 2664 8 of 15

incorporation in the total lipids and phospholipids of GBM neural stem-like cells. FABP7 wasincorporation knocked-down in the total in A4-004N lipids and cells phospho usinglipids lentiviral of GBM shRNA neural constructs stem-like (shFABP7-1cells. FABP7 and Nutrients 2021, 13, 2664 8 of 16 shFABP7-2)was knocked-down (Figure in4). A4-004N In A4-004N cells shControl using lentiviral cells, AAshRNA and constructsDHA treatment (shFABP7-1 increased and the intracellularshFABP7-2) (Figure levels of4). AAIn A4-004N and DHA, shControl respecti cells,vely, AA in and both DHA total treatment lipid and increased total phospho- the lipidintracellular fractions, levels as describedof AA and earlierDHA, respectifor non-transfectedvely, in both totalA4-004N lipid andcells total (Figure phospho- 5). Upon FABP7lipidfractions, fractions, depletion, as described as describedhowever, earlier earlier DHA for non-transfected foruptake non-transfected into total A4-004N lipids A4-004N cells was ( Figurecells significantly (Figure5 ). Upon 5). reduced FABP7Upon in DHA-treatedFABP7depletion, depletion, however, A4-004N however, DHA cells uptake DHA(6.4% into uptakein totalA4-004 lipidsinto shFABP7-1 total was significantlylipids cells was and significantly reduced 6.6% A4-004N in DHA-treated reduced shFABP7- in 2DHA-treatedA4-004N cells, compared cells A4-004N (6.4% to in12.9% cells A4-004 (6.4%in A4-004N shFABP7-1 in A4-004 shCo cells shFABP7-1ntrol and cells) 6.6% cells (Figure A4-004N and 6.6% 5A). shFABP7-2 A4-004N Reduced shFABP7- cells, DHA com- incor- poration2pared cells, tocompared 12.9%also resulted in to A4-004N 12.9% in ain shControl decreased A4-004N cells) shCo DHA:AA (Figurentrol cells) 5ratioA). (Figure Reducedand ω 5A).-3: DHAω -6Reduced ratio incorporation in DHA total incor- lipids also of FABP7-depletedporationresulted inalso a decreasedresulted A4-004N in DHA:AA a decreasedcells compared ratio DHA:AA and ω to-3: A4-004Nω ratio-6 ratio and inshControl ω total-3:ω-6 lipids ratio cells of in FABP7-depleted(Figure total lipids 5C,D). of No- tably,FABP7-depletedA4-004N reduced cells compared DHA A4-004N incorporation to cells A4-004N compared shControlwas tonot A4-004N observed cells (Figure shControl in5 C,D).total cells Notably,phospholipids (Figure reduced 5C,D). of DHA No-FABP7- depletedtably,incorporation reduced cells was (FigureDHA not incorporation observed5E), nor inwere total was the phospholipids not DHA:AA observed and ofin FABP7-depleted totalω-3:ω phospholipids-6 ratios cellsaffected (ofFigure FABP7- by 5FABP7E ), ω ω depletiondepletednor were cells (Figure the DHA:AA(Figure 5G,H). 5E), and Interestingly, nor were-3: -6 the ratios evenDHA:AA affected though and byAA ω FABP7-3: isω also-6 ratios depletiona affected for (Figure FABP7, by FABP75G,H). FABP7 Interestingly, even though AA is also a ligand for FABP7, FABP7 depletion had no signif- depletiondepletion (Figurehad no 5G,H).significant Interestingly, effect on eventhe uptake though of AA AA is inalso either a ligand total for lipids FABP7, (Figure FABP7 5B) or icant effect on the uptake of AA in either total lipids (Figure5B) or total phospholipids totaldepletion phospholipids had no significant (Figure effect 5F). In on agreemen the uptaket withof AA these in either data, total neither lipids the (Figure DHA:AA 5B) or ratio total(Figure phospholipids5F ). In agreement (Figure with5F). In these agreemen data,t neither with these the DHA:AAdata, neither ratio the nor DHA:AA the ω-3: ratioω-6 norratio the was ω-3: affectedω-6 ratio by was FABP7 affected depletion by FABP7 in AA-treated depletion A4-004N in AA-treated cells (Figure A4-004N5C,D,G,H). cells (Fig- urenor the5C,D,G,H). ω-3:ω-6 ratioOur wascombined affected FABP7 by FABP7 results depletion suggest in AA-treatedthe presence A4-004N of a compensatory cells (Fig- ureOur 5C,D,G,H). combined FABP7Our combined results suggest FABP7 the results presence suggest of a compensatorythe presence of mechanism a compensatory for AA mechanism for AA uptake in GBM neural stem-like cells when FABP7 is depleted, and a mechanismuptake in GBM for AA neural uptake stem-like in GBM cells neural when stem-like FABP7 iscells depleted, when FABP7 and a specialized is depleted, role and for a specialized role for FABP7 in the uptake of DHA in total lipids of A4-004N cells. specializedFABP7 in the role uptake for FABP7 of DHA in the in totaluptake lipids of DHA of A4-004N in total cells. lipids of A4-004N cells.

FigureFigureFigure 4. 4. FABP7FABP7 levels levels in inin lentivirus-infected lentivirus-infectedlentivirus-infected A4-004N A4-004N A4-004N cell cells. cells. Westerns. Western Western blot blot blot analysis analysis analysis of of FABP7 FABP7of FABP7 in in total total in total lysateslysateslysates prepared prepared from from A4-004N A4-004NA4-004N ce cells cellslls transfected transfected transfected with with with shControl, shControl, shControl, shFABP7-1 shFABP7-1 shFABP7-1 and and and FABP7-2 FABP7-2 FABP7-2 lentiviral lentiviral lentiviral constructs.constructs.constructs. GAPDHGAPDH GAPDH was was used usedused as asas the thethe loading loading loading control. control. control.

FigureFigure 5. 5. EffectsEffects of of PUFA PUFA treatment on the incorporation of of AA and DHA into total lipids and total phospholipids from from FigureA4-004NA4-004N 5. Effects control of andand PUFA shFABP7shFABP7 treatment knockdown knockdown on the cells. incorporationcells. A4-004N A4-004N shControl, ofshCont AA androl, shFABP7-1, shFABP7-1,DHA into and total and shFABP7-2 lipidsshFABP7-2 and were totalwere cultured phospholipids cultured in medium in me- from A4-004Ndiumsupplemented supplemented control with and BSA withshFABP7 (control), BSA (control), knockdown 30 µM 30 AA, μ Mcells. or AA, 30 A4-004Nµ orM 30 DHA. μM shCont PercentDHA. Percentrol, abundance shFABP7-1, abundance of DHA, and of AA, shFABP7-2DHA, DHA:AA AA, DHA:AA were ratio cultured and ratioω-3: andω in-6 me- diumωratio-3: ωsupplemented in-6 totalratio lipidsin total ( Awith lipids–D) BSA and (A– total(control),D) and phospholipids total 30 phospholipidsμM AA, (E– orH) 30 of ( A4-004NμEM–H DHA.) of A4-004N shControl Percent shControl andabundance shFABP7 and of shFABP7 cells. DHA, **** AA, cells.. indicates DHA:AA **** pindicates< 0.0001.ratio and ωp-3:ns. <ω 0.0001. indicates-6 ratio ns. in notindicates total significant. lipids not (significant.A–D) and total phospholipids (E–H) of A4-004N shControl and shFABP7 cells.. **** indicates p < 0.0001. ns. indicates not significant. FABPsFABPs are are responsible responsible for for both both the the uptake uptake and and trafficking trafficking of of their their fatty fatty acid acid ligands ligands withinwithinFABPs cells. cells. FABP7are FABP7 responsible has has previously previously for both been been the shown shown uptake to to increaseand increase trafficking the the formation formation of their of of lipidfatty lipid droplets,acid droplets, ligands withinan organelle cells. FABP7 responsible has previously for lipid storage been shown [62,63]. to As increase our data the indicate formation that FABP7of lipid expres- droplets, sion increases the uptake of DHA into total lipids, but not phospholipids, we examined whether FABP7 delivers DHA to lipid droplets. We observed an increased accumulation of lipid droplets when A4-004N shControl cells were cultured in medium supplemented

Nutrients 2021, 13, 2664 9 of 15

an organelle responsible for lipid storage [62,63]. As our data indicate that FABP7 expres-

Nutrients 2021, 13, 2664 sion increases the uptake of DHA into total lipids, but not phospholipids, we9 of 16examined whether FABP7 delivers DHA to lipid droplets. We observed an increased accumulation of lipid droplets when A4-004N shControl cells were cultured in medium supplemented with 30 μM DHA (Figure 6). Intriguingly, upon FABP7 depletion, lipid droplet accumu- with 30 µM DHA (Figure6). Intriguingly, upon FABP7 depletion, lipid droplet accumula- tionlation was was significantly significantly reduced, reduced, suggesting suggesting that DHA that may DHA be preferentiallymay be preferentially stored in lipid stored in li- dropletspid droplets in FABP7-expressing in FABP7-expressing cells (Figure cells6 A).(Figur Quantitativee 6A). Quantitative analyses revealed analyses a >2-fold revealed a >2- decreasefold decrease in the in average the average intensity intensity of Nile Redof Nile staining Red staining per cell upon per cell FABP7 upon depletion FABP7 in depletion DHA-treatedin DHA-treated A4-004N A4-004N cells cells (Figure (Figure6B). Similar 6B). Simi resultslar results were observed were observed in U251 cellsin U251 and cells and ED501NED501N cells, cells, both both of of which which also also express express FABP7 FABP7 (Figure (Figure S1) [49 S1)]. [49].

FigureFigure 6. 6.Effect Effect of of DHA DHA treatment treatment on lipidon lipid droplet droplet formation formation in A4-004N in A4-004N shControl shControl and A4-004N and A4-004N shFABP7shFABP7 cells. cells. (A ()A A4-004N) A4-004N shControl shControl and and shFABP7 shFABP7 cells were cells cultured were cultured in medium in medium supplemented supplemented µ withwith BSA BSA control control or or 30 30M μ DHA.M DHA. Cells Cells were were stained stained with Nilewith Red Nile and Red images and images captured captured by confocal by confocal microscopy.microscopy. DAPI DAPI was was used used to stainto stain the nucleus.the nucleus. (B) Quantification (B) Quantification of lipid of droplets lipid droplets in A4-004N in A4-004N shControlshControl and and shFABP7 shFABP7 cells cells cultured cultured in DHA-supplemented in DHA-supplemented medium. Themedium. average The intensity average of Nile intensity of RedNile staining Red staining per cell per was cell measured was measured using raw using images raw (n =images 8, 15–30 (n cells/image) = 8, 15–30 cells/image) taken by confocal taken by con- microscopy.focal microscopy. *** indicates *** indicatesp < 0.001. p < 0.001.

3.4. DHA-Mediated Inhibition of GBM Neural Stem-like Cell Migration Is Dependent on FABP73.4. DHA-Mediated Expression Inhibition of GBM Neural Stem-Like Cell Migration Is Dependent on FABP7We Expression have previously reported that AA promotes, whereas DHA inhibits, U87 GBM cell migrationWe have inpreviously an FABP7-dependent reported that manner AA promotes, [34]. We therefore whereas used DHA the inhibits, Transwell U87 GBM migrationcell migration assay in to examinean FABP7-dependent the effect of DHA manner and FABP7 [34]. expressionWe therefore on GBM used neural the Transwell stem- mi- likegration cell migration.assay to examine A4-004N the shControl effect of cells DHA and and A4-004N FABP7 shFABP7 expression cells on were GBM cultured neural stem- inlike neurosphere cell migration. medium A4-004N supplemented shControl with cells 30 µandM DHA A4-004N or BSA shFABP7 control forcells 24 were h before cultured in carrying out the Transwell assay. As previously shown for adherent GBM cells, migration neurosphere medium supplemented with 30 μM DHA or BSA control for 24 h before car- rates of FABP7-depleted A4-004N cells were significantly reduced compared to A4-004N shControlrying out cells the (FigureTranswell7A,B). assay. DHA treatmentAs previously resulted shown in a >60% for decreaseadherent in GBM the migration cells, migration ofrates A4-004N of FABP7-depleted shControl cells compared A4-004N to cells BSA were control significantly (p < 0.05). In reduced contrast, thecompared migration to of A4-004N A4-004NshControl shFABP7 cells (Figure cells was 7A,B). not affected DHA treatment by DHA treatment. resulted Ourin a combined>60% decrease results in indicate the migration thatof A4-004N although shControl DHA uptake cells is compared observed in to both BSA shControl control ( andp < 0.05). shFABP7 In contrast, cells, it is the only migration whenof A4-004N FABP7 isshFABP7 present that cells uptake was ofnot DHA affected leads by tothe DHA inhibition treatment. of GBM Our neural combined stem-like results in- celldicate migration. that although DHA uptake is observed in both shControl and shFABP7 cells, it is only when FABP7 is present that uptake of DHA leads to the inhibition of GBM neural stem-like cell migration.

Nutrients 2021, 13, 2664 10 of 15 Nutrients 2021, 13, 2664 10 of 16

Figure 7. Effects of DHA treatment on the migration of A4-004N shControl and A4-004N shFABP7 Figurecells. (7.A) Effects Representative of DHA Transwell treatment images on the of A4-004N migration shControl of A4-004N and A4-004N shControl shFABP7 and cellsA4-004N shFABP7 cells.cultured (A) inRepresentative medium supplemented Transwell with images bovine of serum A4- albumin004N shControl (BSA control) and or A4-004N 30 µM DHA. shFABP7 cells cul- tured(B) Quantification in medium of migratingsupplemented cells described with inbovine (A). Differences serum werealbumin assessed (BSA for significancecontrol) or using 30 μM DHA. (B) Quantificationthe two-tailed unpaired of migratingt-test. n =cells 3. *** described indicates p in< 0.005. (A). Differences were assessed for significance using the two-tailed unpaired t-test. n = 3. *** indicates p < 0.005 4. Discussion 4. DiscussionGBM tumors share key features with the development of the central nervous system (CNS) [64]. First, GBM tumors have subpopulations of neural stem-like cells that express neuralGBM stem tumors cell markers, share such key asfeatures Nestin, with CD133, the and de FABP7.velopment Second, of the migration central and nervous system (CNS)infiltration [64]. of First, GBM GBM cells share tumors morphologically have subpopulations and structurally of similarneural features stem-like to those cells that express neuralassociated stem with cell long-distance markers, such migration as Nestin, during CD133, and in FABP7. developing Second, brain. Third, migration and in- the DHA:AA ratio in GBM tissue is similar to that seen in the fetal brain when neural filtrationcells are undergoingof GBM cells long share distance morphologically migrations [30,31]. and Notably, structurally FABP7 is similar essential features for to those associatedboth the maintenance with long-distance of radial glial migration progenitor during cells [ 65neurogenesis] and neuronal in cell developing migration brain. Third, theduring DHA:AA brain development ratio in GBM [38,39 tissue]. In standard is similar GBM to adherent that seen cell in cultures, the fetal overexpression brain when neural cells areof FABP7undergoing increases, long whereas distance knockdown migrations of FABP7 [30,31 decreases]. Notably, GBM FABP7 cell migration is essential and for both the infiltration [16,42]. Moreover, FABP7 is highly expressed in GBM neural stem-like cells [40] maintenanceand its elevated of levels radial are associatedglial progenitor with poor cells clinical [65] outcome and neuronal [62]. In combination, cell migration these during brain developmentdata suggest that [38,39]. FABP7 mayIn standard be able to hijackGBM the adhere brain’snt normal cell cultures, developmental overexpression processes of FABP7 increases,for the maintenance whereas of knockdown GBM stemness of and FABP7 tumor infiltrationdecreases in GBM brain parenchyma.cell migration and infiltration 0 [16,42].The Moreover, brain is highly FABP7 enriched is in highly long chain expressed PUFAs, especially in GBM FABP7 neurals preferred stem-like ligands, cells [40] and its ω ω elevated-3 DHA levels and are-6 AA, associated which make with up poor 8% and clinical 6% of outcome the dry weight [62]. of In adult combination, human these data brain, respectively [29,66]. FABP7 is found in both the cytoplasm and nucleus, as well as at suggestthe plasma that membrane FABP7 may where be it isable involved to hijack in the th uptakee brain’s of PUFAs normal [34 ,develo49]. FABP7pmental plays processes for thedifferent maintenance roles in the of cell GBM depending stemness on whichand tumo ligandr itinfiltration is bound to in [34 brain,66,67]. parenchyma. DHA has previouslyThe brain been shownis highly to inhibit enriched the migration in long chain of FABP7-expressing PUFAs, especially GBM cells FABP7 [34,35′s], preferred lig- ands,as well ω as-3 sensitizeDHA and GBM ω-6 cells AA, to which chemotherapy make up and 8% radiation and 6%in vitroof the[32 dry,37, 68weight,69]. It of is adult human already known that dietary DHA can affect the fatty acid composition of lipids in brain brain, respectively [29,66]. FABP7 is found in both the cytoplasm and nucleus, as well as at the plasma membrane where it is involved in the uptake of PUFAs [34,49]. FABP7 plays different roles in the cell depending on which ligand it is bound to [34,66,67]. DHA has previously been shown to inhibit the migration of FABP7-expressing GBM cells [34,35], as well as sensitize GBM cells to chemotherapy and radiation in vitro [32,37,68,69]. It is al- ready known that dietary DHA can affect the fatty acid composition of lipids in brain tissue [70–72], opening the door to the possibility that GBM patient outcome could be im- proved by increasing the DHA content in the tumor microenvironment.

Nutrients 2021, 13, 2664 11 of 16

tissue [70–72], opening the door to the possibility that GBM patient outcome could be improved by increasing the DHA content in the tumor microenvironment. Neural stem-like cells are key players in GBM cellular heterogeneity and therapy resis- tance [46,73]. expression profiling has revealed significant differences between neural stem-like GBM cells cultured under neurosphere conditions and adherent GBM cells cul- tured in serum-containing medium, with the neural stem-like cells more closely mirroring the original tumor [47,48,74]. Furthermore, compared to their matched serum-differentiated counterparts, GBM neurosphere cultures are enriched in enzymes involved in the PUFA synthesis cascade such as ELOVL2 and FASD2 [19]. Cyclooxygenase 2 (COX-2), which metabolizes AA to its downstream bioactive metabolites (e.g., prostaglandins), is prefer- entially activated in GBM neural stem-like cells compared to adherent cells [75]. We have previously shown that COX-2 is upregulated in FABP7-expressing GBM cells cultured in AA-rich medium [34]. Interestingly, when patient-derived GBM tumors are sorted into fast- versus slow-cycling (neural stem-like) cells, the latter not only preferentially express FABP7 but have elevated levels of PUFA metabolism intermediates [62]. Thus, there is an emerging link between stemness, FABP7 expression, and increased PUFA metabolism in GBM. PUFA metabolism is gaining recognition as an important contributor to GBM tumori- genic properties. However, little is known about the uptake and trafficking of PUFAs in GBM neural stem-like cells. To date, the effect of DHA on GBM fatty acid composition has only been investigated using established GBM cell lines cultured under adherent (differentiation-promoting) conditions [32]. By comparing the fatty acid composition of patient-derived GBM neural stem-like cells with that of their adherent counterparts, we found that DHA treatment effectively increases the DHA content in both total lipids and total phospholipids of GBM neural stem-like cells, but especially in total lipids. Our results further indicate that FABP7 plays an important role in the efficient uptake of DHA in total lipids but not phospholipids. This preferential link between DHA, FABP7, and total lipids is particularly interesting in light of our recent finding that DHA treatment disrupts FABP7 nanodomains clustered on the surface of GBM neural stem-like cells and promotes FABP7 localization to mitochondria [49]. Our previous work also shows that DHA treatment in FABP7-expressing GBM cells promotes the nuclear localization of FABP7 and induction of PPAR activity [34]. Thus, FABP7’s main role in DHA-supplemented GBM cells may be to facilitate the uptake, intracellular transport, and utilization of DHA for functions that are unrelated or not directly related to phospholipids. Along with the decrease in DHA uptake in total lipids observed in FABP7-depleted GBM neural stem like cells, we also found that FABP7 depletion decreased the formation of lipid droplets in GBM neural stem-like cells cultured in DHA-supplemented medium. Associations between FABP7 expression and lipid droplet formation have been previously reported. For example, FABP7 expression is associated with elevated numbers of lipid droplets in both astrocytes and GBM cells [62,63]. Furthermore, up-regulation of FABP7 expression in U87 adherent GBM cells cultured under hypoxic conditions was accompanied by increased fatty acid uptake and increased formation of lipid droplets [76]. Long believed to simply serve as storage sites for fats, lipid droplets are now known to be hubs that coordinate a wide range of lipid-related functions ranging from delivery of fatty acids to mitochondria, regulation of membrane dynamics, and timed release of bioactive lipids that regulate inflammation [77,78]. DHA has already been implicated in the remodeling of lipid droplets in microglia, with a demonstrated effect on the inhibition of neuro- inflammation [79]. Based on these combined data, one may therefore postulate that FABP7 expression in GBM neural stem-like cells has the potential of inducing an anti-tumorigenic response when cells are cultured in a DHA-rich microenvironment that promotes the formation of DHA-rich lipid droplets. Consistent with the idea that DHA has anti-tumorigenic properties in FABP7-expressing GBM neural stem-like cells, we found that DHA inhibited the migration of FABP7-expressing GBM neural stem-like cells but had negligible effects on their FABP7-depleted counterparts. These results are consistent with our previous findings using the established adherent U87 Nutrients 2021, 13, 2664 12 of 16

GBM cell line [34] and indicate that the migratory properties of FABP7-expressing GBM cells are dependent on the ratio of DHA:AA in the tumor microenvironment. Together, our findings suggest that infiltrative FABP7-expressing GBM neural stem-like cells will be selectively targeted by DHA treatment. Thus, while expression of FABP7 may promote GBM growth in an AA-rich tumor microenvironment, in a DHA-rich microenvironment, FABP7 may inhibit tumor infiltration as the result of increased DHA uptake and utilization. We thus propose that FABP7 is the Achilles’ heel of GBM neural stem-like cells, with the potential of inhibiting the migration/infiltration of these cells in a DHA-rich microenviron- ment. DHA-rich diets have already been shown to inhibit breast cancer xenograft tumor growth and metastasis [80,81], delay neuroblastoma cancer progression in immunodefi- cient mice [82], as well as increase the in vitro and in vivo efficacy of chemotherapy drugs used for the treatment of colon cancer [83–86]. It will be important to investigate whether DHA supplementation can increase DHA levels in both the GBM microenvironment and GBM tissue, thereby paving the way to improved GBM patient outcome, by inhibiting the infiltration of FABP7-expressing GBM neural stem-like cells into brain parenchyma.

Supplementary Materials: The following are available online at https://www.mdpi.com/article/ 10.3390/nu13082664/s1, Figure S1: Effects of DHA treatment on lipid droplet formation in U251 and ED501N cells with or without FABP7 depletion, Table S1: Fatty acid composition of total lipids extracted from A4-004N and A4-004Adh cells, Table S2: Fatty acid composition of total lipids extracted from A4-004N and A4-004Adh cells cultured in media supplemented with BSA (control), 30 µM AA or 30 µM DHA, Table S3: Fatty acid composition of total lipids and total phospholipids from A4-004N and A4-004Adh cells cultured under normal culture conditions and in media supplemented with 30 µM BSA, Table S4: Fatty acid composition of total phospholipids from A4-004N and A4-004Adh cells, Table S5: Fatty acid composition of total phospholipids from A4-004N and A4-004Adh cells cultured in media supplemented with BSA (control), 30 µM AA or 30 µM DHA. Author Contributions: W.-S.C. and X.X. made equal contributions to the manuscript. Conceptualiza- tion, W.-S.C., X.X., C.J.F. and R.G.; methodology, W.-S.C., X.X., S.G. and Y.W.; formal analysis, W.-S.C., X.X., S.G. and Y.W.; investigation, W.-S.C., X.X., S.P., M.C., C.J.F. and R.G.; resources, S.P., M.C.; writing—original draft preparation, W.-S.C., X.X. and R.G.; writing—review and editing, W.-S.C., X.X., S.G., Y.W., S.P., M.C., C.J.F. and R.G.; visualization, W.-S.C. and X.X.; supervision, C.J.F. and R.G.; project administration, R.G.; funding acquisition, R.G. All authors have read and approved the paper. All authors have read and agreed to the published version of the manuscript. Funding: This work was supported by a grant to Roseline Godbout from the Canadian Institutes of Health Research (CIHR)—Funding Reference Number 130314. Institutional Review Board Statement: Not applicable. Informed Consent Statement: Not applicable. Data Availability Statement: No new data were created or analyzed in this study. Data sharing is not applicable to this article. Acknowledgments: We thank Liz Monckton for technical assistance. We are grateful to Hua Chen and Kenneth Petruk for the ED501 GBM cell line. Conflicts of Interest: The authors declare no conflict of interest.

References 1. Wen, P.Y.; Kesari, S. Malignant gliomas in adults. N. Engl. J. Med. 2008, 359, 492–507. [CrossRef][PubMed] 2. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2012–2016. Neuro Oncol. 2019, 21, v1–v100. [CrossRef][PubMed] 3. Buckner, J.C. Factors influencing survival in high-grade gliomas. Semin. Oncol. 2003, 30, 10–14. [CrossRef] 4. Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [CrossRef] Nutrients 2021, 13, 2664 13 of 16

5. Vehlow, A.; Cordes, N. Invasion as target for therapy of glioblastoma multiforme. Biochim. Biophys. Acta 2013, 1836, 236–244. [CrossRef] 6. Aum, D.J.; Kim, D.H.; Beaumont, T.L.; Leuthardt, E.C.; Dunn, G.P.; Kim, A.H. Molecular and cellular heterogeneity: The hallmark of glioblastoma. Neurosurg. Focus 2014, 37, E11. [CrossRef] 7. Bonavia, R.; Inda, M.M.; Cavenee, W.K.; Furnari, F.B. Heterogeneity maintenance in glioblastoma: A social network. Cancer Res. 2011, 71, 4055–4060. [CrossRef][PubMed] 8. Yi, Y.; Hsieh, I.Y.; Huang, X.; Li, J.; Zhao, W. Glioblastoma Stem-like Cells: Characteristics, Microenvironment, and Therapy. Front Pharmacol. 2016, 7, 477. [CrossRef][PubMed] 9. Lathia, J.D.; Mack, S.C.; Mulkearns-Hubert, E.E.; Valentim, C.L.; Rich, J.N. Cancer stem cells in glioblastoma. Genes Dev. 2015, 29, 1203–1217. [CrossRef] 10. Pallini, R.; Ricci-Vitiani, L.; Banna, G.L.; Signore, M.; Lombardi, D.; Todaro, M.; Stassi, G.; Martini, M.; Maira, G.; Larocca, L.M.; et al. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin. Cancer Res. 2008, 14, 8205–8212. [CrossRef] 11. Eramo, A.; Ricci-Vitiani, L.; Zeuner, A.; Pallini, R.; Lotti, F.; Sette, G.; Pilozzi, E.; Larocca, L.M.; Peschle, C.; De Maria, R. Chemotherapy resistance of glioblastoma stem cells. Cell Death Differ. 2006, 13, 1238–1241. [CrossRef][PubMed] 12. Vlashi, E.; Lagadec, C.; Vergnes, L.; Matsutani, T.; Masui, K.; Poulou, M.; Popescu, R.; Della Donna, L.; Evers, P.; Dekmezian, C.; et al. Metabolic state of glioma stem cells and nontumorigenic cells. Proc. Natl. Acad. Sci. USA 2011, 108, 16062–16067. [CrossRef][PubMed] 13. Yasumoto, Y.; Miyazaki, H.; Vaidyan, L.K.; Kagawa, Y.; Ebrahimi, M.; Yamamoto, Y.; Ogata, M.; Katsuyama, Y.; Sadahiro, H.; Suzuki, M.; et al. Inhibition of Fatty Acid Synthase Decreases Expression of Stemness Markers in Glioma Stem Cells. PLoS ONE 2016, 11, e0147717. [CrossRef] 14. Strickland, M.; Stoll, E.A. Metabolic Reprogramming in Glioma. Front Cell Dev. Biol. 2017, 5, 43. [CrossRef][PubMed] 15. Garnier, D.; Renoult, O.; Alves-Guerra, M.C.; Paris, F.; Pecqueur, C. Glioblastoma Stem-like Cells, Metabolic Strategy to Kill a Challenging Target. Front Oncol. 2019, 9, 118. [CrossRef] 16. De Rosa, A.; Pellegatta, S.; Rossi, M.; Tunici, P.; Magnoni, L.; Speranza, M.C.; Malusa, F.; Miragliotta, V.; Mori, E.; Finocchiaro, G.; et al. A Radial Glia Gene Marker, Fatty Acid Binding Protein 7 (FABP7), Is Involved in Proliferation and Invasion of Glioblastoma Cells. PLoS ONE 2012, 7, e52113. [CrossRef] 17. Hale, J.S.; Otvos, B.; Sinyuk, M.; Alvarado, A.G.; Hitomi, M.; Stoltz, K.; Wu, Q.; Flavahan, W.; Levison, B.; Johansen, M.L.; et al. Cancer stem cell-specific scavenger receptor CD36 drives glioblastoma progression. Stem Cells 2014, 32, 1746–1758. [CrossRef] [PubMed] 18. Duman, C.; Yaqubi, K.; Hoffmann, A.; Acikgoz, A.A.; Korshunov, A.; Bendszus, M.; Herold-Mende, C.; Liu, H.K.; Alfonso, J. Acyl- CoA-Binding Protein Drives Glioblastoma Tumorigenesis by Sustaining Fatty Acid Oxidation. Cell Metab. 2019, 30, 274–289.e275. [CrossRef] 19. Gimple, R.C.; Kidwell, R.L.; Kim, L.J.Y.; Sun, T.; Gromovsky, A.D.; Wu, Q.; Wolf, M.; Lv, D.; Bhargava, S.; Jiang, L.; et al. Glioma Stem Cell-Specific Superenhancer Promotes Polyunsaturated Fatty-Acid Synthesis to Support EGFR Signaling. Cancer Discov. 2019, 9, 1248–1267. [CrossRef] 20. Bazinet, R.P.; Laye, S. Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat. Rev. Neurosci. 2014, 15, 771–785. [CrossRef] 21. Lacombe, R.J.S.; Chouinard-Watkins, R.; Bazinet, R.P. Brain docosahexaenoic acid uptake and metabolism. Mol. Aspects Med. 2018, 64, 109–134. [CrossRef] 22. Azrad, M.; Turgeon, C.; Demark-Wahnefried, W. Current evidence linking polyunsaturated Fatty acids with cancer risk and progression. Front. Oncol. 2013, 3, 224. [CrossRef][PubMed] 23. Ricciotti, E.; FitzGerald, G.A. Prostaglandins and inflammation. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 986–1000. [CrossRef] [PubMed] 24. Qiu, J.; Shi, Z.; Jiang, J. Cyclooxygenase-2 in glioblastoma multiforme. Drug. Discov. Today 2017, 22, 148–156. [CrossRef][PubMed] 25. Rand, A.A.; Barnych, B.; Morisseau, C.; Cajka, T.; Lee, K.S.S.; Panigrahy, D.; Hammock, B.D. Cyclooxygenase-derived proangio- genic metabolites of epoxyeicosatrienoic acids. Proc. Natl. Acad. Sci. USA 2017, 114, 4370–4375. [CrossRef] 26. Serhan, C.N.; Levy, B.D. Resolvins in inflammation: Emergence of the pro-resolving superfamily of mediators. J. Clin. Investig. 2018, 128, 2657–2669. [CrossRef] 27. Sulciner, M.L.; Serhan, C.N.; Gilligan, M.M.; Mudge, D.K.; Chang, J.; Gartung, A.; Lehner, K.A.; Bielenberg, D.R.; Schmidt, B.; Dalli, J.; et al. Resolvins suppress tumor growth and enhance cancer therapy. J. Exp. Med. 2018, 215, 115–140. [CrossRef] 28. Zhang, Q.; Zhu, B.; Li, Y. Resolution of Cancer-Promoting Inflammation: A New Approach for Anticancer Therapy. Front. Immunol. 2017, 8, 71. [CrossRef] 29. Owada, Y. Fatty acid binding protein: Localization and functional significance in the brain. Tohoku J. Exp. Med. 2008, 214, 213–220. [CrossRef] 30. Martin, D.D.; Robbins, M.E.; Spector, A.A.; Wen, B.C.; Hussey, D.H. The fatty acid composition of human gliomas differs from that found in nonmalignant brain tissue. Lipids 1996, 31, 1283–1288. [CrossRef] 31. Marszalek, R.; Pisklak, M.; Jankowski, W.; Lukaszkiewicz, J.; Horsztynski, D.; Wawer, I. NMR and gas chromatography studies of lyophilized human brain tumors. Acta Pol. Pharm. 2010, 67, 129–136. [PubMed] Nutrients 2021, 13, 2664 14 of 16

32. Harvey, K.A.; Xu, Z.; Saaddatzadeh, M.R.; Wang, H.; Pollok, K.; Cohen-Gadol, A.A.; Siddiqui, R.A. Enhanced anticancer properties of lomustine in conjunction with docosahexaenoic acid in glioblastoma cell lines. J. Neurosurg. 2015, 122, 547–556. [CrossRef] 33. Leonardi, F.; Attorri, L.; Di Benedetto, R.; Di Biase, A.; Sanchez, M.; Nardini, M.; Salvati, S. Effect of arachidonic, eicosapentaenoic and docosahexaenoic acids on the oxidative status of C6 glioma cells. Free Radic. Res. 2005, 39, 865–874. [CrossRef][PubMed] 34. Mita, R.; Beaulieu, M.J.; Field, C.; Godbout, R. Brain fatty acid-binding protein and omega-3/omega-6 fatty acids: Mechanistic insight into malignant glioma cell migration. J. Biol. Chem. 2010, 285, 37005–37015. [CrossRef][PubMed] 35. Ruan, M.; Liu, J.; Ren, X.; Li, C.; Zhao, A.Z.; Li, L.; Yang, H.; Dai, Y.; Wang, Y. Whole transcriptome sequencing analyses of DHA treated glioblastoma cells. J. Neurol. Sci. 2019, 396, 247–253. [CrossRef][PubMed] 36. Kim, S.; Jing, K.; Shin, S.; Jeong, S.; Han, S.H.; Oh, H.; Yoo, Y.S.; Han, J.; Jeon, Y.J.; Heo, J.Y.; et al. omega3-polyunsaturated fatty acids induce cell death through apoptosis and autophagy in glioblastoma cells: In vitro and in vivo. Oncol. Rep. 2018, 39, 239–246. 37. Antal, O.; Hackler, L., Jr.; Shen, J.; Man, I.; Hideghety, K.; Kitajka, K.; Puskas, L.G. Combination of unsaturated fatty acids and ionizing radiation on human glioma cells: Cellular, biochemical and analysis. Lipids Health Dis. 2014, 13, 142. [CrossRef] 38. Feng, L.; Hatten, M.E.; Heintz, N. Brain lipid-binding protein (BLBP): A novel signaling system in the developing mammalian CNS. Neuron 1994, 12, 895–908. [CrossRef] 39. Kurtz, A.; Zimmer, A.; Schnutgen, F.; Bruning, G.; Spener, F.; Muller, T. The expression pattern of a novel gene encoding brain-fatty acid binding protein correlates with neuronal and glial cell development. Development 1994, 120, 2637–2649. [CrossRef] 40. Morihiro, Y.; Yasumoto, Y.; Vaidyan, L.K.; Sadahiro, H.; Uchida, T.; Inamura, A.; Sharifi, K.; Ideguchi, M.; Nomura, S.; Tokuda, N.; et al. Fatty acid binding protein 7 as a marker of glioma stem cells. Pathol. Int. 2013, 63, 546–553. [CrossRef] 41. Balendiran, G.K.; Schnutgen, F.; Scapin, G.; Borchers, T.; Xhong, N.; Lim, K.; Godbout, R.; Spener, F.; Sacchettini, J.C. Crystal structure and thermodynamic analysis of human brain fatty acid-binding protein. J. Biol. Chem. 2000, 275, 27045–27054. [CrossRef] 42. Mita, R.; Coles, J.E.; Glubrecht, D.D.; Sung, R.; Sun, X.; Godbout, R. B-FABP-expressing radial glial cells: The malignant glioma cell of origin? Neoplasia 2007, 9, 734–744. [CrossRef][PubMed] 43. Liang, Y.; Diehn, M.; Watson, N.; Bollen, A.W.; Aldape, K.D.; Nicholas, M.K.; Lamborn, K.R.; Berger, M.S.; Botstein, D.; Brown, P.O.; et al. Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. Proc. Natl. Acad. Sci. USA 2005, 102, 5814–5819. [CrossRef][PubMed] 44. Nasrollahzadeh, J.; Siassi, F.; Doosti, M.; Eshraghian, M.R.; Shokri, F.; Modarressi, M.H.; Mohammadi-Asl, J.; Abdi, K.; Nikmanesh, A.; Karimian, S.M. The influence of feeding linoleic, gamma-linolenic and docosahexaenoic acid rich oils on rat brain tumor fatty acids composition and fatty acid binding protein 7 mRNA expression. Lipids Health Dis. 2008, 7, 45. [CrossRef][PubMed] 45. Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [CrossRef] 46. Osuka, S.; Van Meir, E.G. Overcoming therapeutic resistance in glioblastoma: The way forward. J. Clin. Investig. 2017, 127, 415–426. [CrossRef] 47. Lenting, K.; Verhaak, R.; Ter Laan, M.; Wesseling, P.; Leenders, W. Glioma: Experimental models and reality. Acta Neuropathol. 2017, 133, 263–282. [CrossRef] 48. Lee, J.; Kotliarova, S.; Kotliarov, Y.; Li, A.; Su, Q.; Donin, N.M.; Pastorino, S.; Purow, B.W.; Christopher, N.; Zhang, W.; et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006, 9, 391–403. [CrossRef] 49. Xu, X.; Wang, Y.; Choi, W.S.; Sun, X.; Godbout, R. Super resolution microscopy reveals DHA-dependent alterations in glioblastoma membrane remodelling and cell migration. Nanoscale 2021, 13, 9706–9722. [CrossRef] 50. Layne, K.S.; Goh, Y.K.; Jumpsen, J.A.; Ryan, E.A.; Chow, P.; Clandinin, M.T. Normal subjects consuming physiological levels of 18:3(n-3) and 20:5(n-3) from flaxseed or fish oils have characteristic differences in plasma lipid and lipoprotein fatty acid levels. J. Nutr. 1996, 126, 2130–2140. [CrossRef][PubMed] 51. Cruz-Hernandez, C.; Goeuriot, S.; Giuffrida, F.; Thakkar, S.K.; Destaillats, F. Direct quantification of fatty acids in human milk by gas chromatography. J. Chromatogr. A 2013, 1284, 174–179. [CrossRef] 52. Brun, M.; Jain, S.; Monckton, E.A.; Godbout, R. Nuclear Factor I Represses the Notch Effector HEY1 in Glioblastoma. Neoplasia 2018, 20, 1023–1037. [CrossRef] 53. Saurty-Seerunghen, M.S.; Bellenger, L.; El-Habr, E.A.; Delaunay, V.; Garnier, D.; Chneiweiss, H.; Antoniewski, C.; Morvan-Dubois, G.; Junier, M.P. Capture at the single cell level of metabolic modules distinguishing aggressive and indolent glioblastoma cells. Acta Neuropathol. Commun. 2019, 7, 155. [CrossRef][PubMed] 54. Lands, W.E. Metabolism of glycerolipides; a comparison of lecithin and triglyceride synthesis. J. Biol. Chem. 1958, 231, 883–888. [CrossRef] 55. Fuentes, N.R.; Kim, E.; Fan, Y.Y.; Chapkin, R.S. Omega-3 fatty acids, membrane remodeling and cancer prevention. Mol. Aspects Med. 2018, 64, 79–91. [CrossRef][PubMed] 56. Levental, K.R.; Malmberg, E.; Symons, J.L.; Fan, Y.Y.; Chapkin, R.S.; Ernst, R.; Levental, I. Lipidomic and biophysical homeostasis of mammalian membranes counteracts dietary lipid perturbations to maintain cellular fitness. Nat. Commun. 2020, 11, 1339. [CrossRef][PubMed] Nutrients 2021, 13, 2664 15 of 16

57. Fuentes, N.R.; Mlih, M.; Barhoumi, R.; Fan, Y.Y.; Hardin, P.; Steele, T.J.; Behmer, S.; Prior, I.A.; Karpac, J.; Chapkin, R.S. Long- Chain n-3 Fatty Acids Attenuate Oncogenic KRas-Driven Proliferation by Altering Plasma Membrane Nanoscale Proteolipid Composition. Cancer Res. 2018, 78, 3899–3912. [CrossRef] 58. Corsetto, P.A.; Montorfano, G.; Zava, S.; Jovenitti, I.E.; Cremona, A.; Berra, B.; Rizzo, A.M. Effects of n-3 PUFAs on breast cancer cells through their incorporation in plasma membrane. Lipids Health Dis. 2011, 10, 73. [CrossRef][PubMed] 59. Furuhashi, M.; Hotamisligil, G.S. Fatty acid-binding proteins: Role in metabolic diseases and potential as drug targets. Nat. Rev. Drug Discov. 2008, 7, 489–503. [CrossRef] 60. Amiri, M.; Yousefnia, S.; Seyed Forootan, F.; Peymani, M.; Ghaedi, K.; Nasr Esfahani, M.H. Diverse roles of fatty acid binding proteins (FABPs) in development and pathogenesis of cancers. Gene 2018, 676, 171–183. [CrossRef] 61. Liu, R.Z.; Mita, R.; Beaulieu, M.; Gao, Z.; Godbout, R. Fatty acid binding proteins in brain development and disease. Int. J. Dev. Biol. 2010, 54, 1229–1239. [CrossRef][PubMed] 62. Hoang-Minh, L.B.; Siebzehnrubl, F.A.; Yang, C.; Suzuki-Hatano, S.; Dajac, K.; Loche, T.; Andrews, N.; Schmoll Massari, M.; Patel, J.; Amin, K.; et al. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J. 2018, 37, e98772. [CrossRef] 63. Islam, A.; Kagawa, Y.; Miyazaki, H.; Shil, S.K.; Umaru, B.A.; Yasumoto, Y.; Yamamoto, Y.; Owada, Y. FABP7 Protects Astrocytes Against ROS Toxicity via Lipid Droplet Formation. Mol. Neurobiol. 2019, 56, 5763–5779. [CrossRef] 64. Jung, E.; Alfonso, J.; Osswald, M.; Monyer, H.; Wick, W.; Winkler, F. Emerging intersections between neuroscience and glioma biology. Nat. Neurosci. 2019, 22, 1951–1960. [CrossRef] 65. Arai, Y.; Funatsu, N.; Numayama-Tsuruta, K.; Nomura, T.; Nakamura, S.; Osumi, N. Role of Fabp7, a downstream gene of Pax6, in the maintenance of neuroepithelial cells during early embryonic development of the rat cortex. J. Neurosci. 2005, 25, 9752–9761. [CrossRef][PubMed] 66. Elsherbiny, M.E.; Emara, M.; Godbout, R. Interaction of brain fatty acid-binding protein with the polyunsaturated fatty acid environment as a potential determinant of poor prognosis in malignant glioma. Prog. Lipid Res. 2013, 52, 562–570. [CrossRef] 67. Kagawa, Y.; Umaru, B.A.; Ariful, I.; Shil, S.K.; Miyazaki, H.; Yamamoto, Y.; Ogata, M.; Owada, Y. Role of FABP7 in tumor cell signaling. Adv. Biol. Regul. 2019, 71, 206–218. [CrossRef][PubMed] 68. Wang, F.; Bhat, K.; Doucette, M.; Zhou, S.; Gu, Y.; Law, B.; Liu, X.; Wong, E.T.; Kang, J.X.; Hsieh, T.C.; et al. Docosahexaenoic acid (DHA) sensitizes brain tumor cells to etoposide-induced apoptosis. Curr. Mol. Med. 2011, 11, 503–511. [CrossRef] 69. Manda, K.; Kriesen, S.; Hildebrandt, G.; Fietkau, R.; Klautke, G. Omega-3 fatty acid supplementation in cancer therapy: Does eicosapentanoic acid influence the radiosensitivity of tumor cells? Strahlenther Onkol. 2011, 187, 127–134. [CrossRef] 70. Elsherbiny, M.E.; Goruk, S.; Monckton, E.A.; Richard, C.; Brun, M.; Emara, M.; Field, C.J.; Godbout, R. Long-Term Effect of Docosahexaenoic Acid Feeding on Lipid Composition and Brain Fatty Acid-Binding Protein Expression in Rats. Nutrients 2015, 7, 8802–8817. [CrossRef] 71. Lauritzen, L.; Brambilla, P.; Mazzocchi, A.; Harslof, L.B.; Ciappolino, V.; Agostoni, C. DHA Effects in Brain Development and Function. Nutrients 2016, 8, 6. [CrossRef][PubMed] 72. Di Nicolantonio, J.J.O.K.; James, H. The Importance of Marine Omega-3s for Brain Development and the Prevention and Treatment of Behavior, Mood, and Other Brain Disorders. Nutrients 2020, 12, 2333. [CrossRef] 73. Prager, B.C.; Bhargava, S.; Mahadev, V.; Hubert, C.G.; Rich, J.N. Glioblastoma Stem Cells: Driving Resilience through Chaos. Trends Cancer 2020, 6, 223–235. [CrossRef][PubMed] 74. Kim, E.J.; Jin, X.; Kim, O.R.; Ham, S.W.; Park, S.H.; Kim, H. Glioma stem cells and their non-stem differentiated glioma cells exhibit differences in mitochondrial structure and function. Oncol. Rep. 2018, 39, 411–416. [CrossRef] 75. Wu, M.; Guan, J.; Li, C.; Gunter, S.; Nusrat, L.; Ng, S.; Dhand, K.; Morshead, C.; Kim, A.; Das, S. Aberrantly activated Cox-2 and Wnt signaling interact to maintain cancer stem cells in glioblastoma. Oncotarget 2017, 8, 82217–82230. [CrossRef] 76. Bensaad, K.; Favaro, E.; Lewis, C.A.; Peck, B.; Lord, S.; Collins, J.M.; Pinnick, K.E.; Wigfield, S.; Buffa, F.M.; Li, J.L.; et al. Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014, 9, 349–365. [CrossRef] 77. Petan, T.; Jarc, E.; Jusovic, M. Lipid Droplets in Cancer: Guardians of Fat in a Stressful World. Molecules 2018, 23, 1941. [CrossRef] 78. Bozza PT, Bakker-Abreu I, Navarro-Xavier RA, Bandeira-Melo C: Lipid body function in eicosanoid synthesis: An update. Prostaglandins Leukot. Essent. Fatty Acids 2011, 85, 205–213. [CrossRef] 79. Tremblay, M.E.; Zhang, I.; Bisht, K.; Savage, J.C.; Lecours, C.; Parent, M.; Titorenko, V.; Maysinger, D. Remodeling of lipid bodies by docosahexaenoic acid in activated microglial cells. J. Neuroinflamm. 2016, 13, 116. [CrossRef][PubMed] 80. Newell, M.; Goruk, S.; Mazurak, V.; Postovit, L.; Field, C.J. Role of docosahexaenoic acid in enhancement of docetaxel action in patient-derived breast cancer xenografts. Breast Cancer Res. Treat. 2019, 177, 357–367. [CrossRef] 81. Yun, E.J.; Song, K.S.; Shin, S.; Kim, S.; Heo, J.Y.; Kweon, G.R.; Wu, T.; Park, J.I.; Lim, K. Docosahexaenoic acid suppresses breast cancer cell metastasis by targeting matrix-metalloproteinases. Oncotarget 2016, 7, 49961–49971. [CrossRef][PubMed] 82. Barnes, C.M.; Prox, D.; Christison-Lagay, E.A.; Le, H.D.; Short, S.; Cassiola, F.; Panigrahy, D.; Chaponis, D.; Butterfield, C.; Nehra, D.; et al. Inhibition of neuroblastoma cell proliferation with omega-3 fatty acids and treatment of a murine model of human neuroblastoma using a diet enriched with omega-3 fatty acids in combination with sunitinib. Pediatr. Res. 2012, 71, 168–178. [CrossRef][PubMed] Nutrients 2021, 13, 2664 16 of 16

83. Vasudevan, A.; Yu, Y.; Banerjee, S.; Woods, J.; Farhana, L.; Rajendra, S.G.; Patel, A.; Dyson, G.; Levi, E.; Maddipati, K.R.; et al. Omega-3 fatty acid is a potential preventive agent for recurrent colon cancer. Cancer Prev. Res. 2014, 7, 1138–1148. [CrossRef] 84. Jordan, A.; Stein, J. Effect of an omega-3 fatty acid containing lipid emulsion alone and in combination with 5-fluorouracil (5-FU) on growth of the colon cancer cell line Caco-2. Eur. J. Nutr. 2003, 42, 324–331. [CrossRef][PubMed] 85. Rani, I.; Vaiphei, K.; Agnihotri, N. Supplementation of fish oil augments efficacy and attenuates toxicity of 5-fluorouracil in 1,2-dimethylhydrazine dihydrochloride/dextran sulfate sodium-induced colon carcinogenesis. Cancer Chemother. Pharmacol. 2014, 74, 309–322. [CrossRef][PubMed] 86. Calviello, G.; Di Nicuolo, F.; Serini, S.; Piccioni, E.; Boninsegna, A.; Maggiano, N.; Ranelletti, F.O.; Palozza, P. Docosahexaenoic acid enhances the susceptibility of human colorectal cancer cells to 5-fluorouracil. Cancer Chemother. Pharmacol. 2005, 55, 12–20. [CrossRef]