<<

Supporting Information

Khaliullina et al. 10.1073/pnas.1416463112 SI Materials and Methods 10 min. The supernatant containing hemolymph was centri- Mammalian Cell Culture. HeLa cells were passaged in DMEM with fuged at 16,000 × g for 2 h to remove the debris and subjected to 10% FBS (Gibco), 50 U/mL penicillin, and 50 μg/mL strepto- lipid extraction. mycin (Gibco). ShhLIGHT2 cells, which are NIH 3T3 cells that express firefly Lipid Extraction. Lipids from purified VLDL, LDL, HDL, and Lpp luciferase under the control of a Gli-responsive promoter, and particles applied as crude extracts in signaling assays were Renilla luciferase controlled by the viral Thymidine kinase promoter extracted by a two-step Bligh and Dyer method (10) and applied (1) were maintained in DMEM + 10% (vol/vol) FBS, 150 μg/mL to ShhLIGHT2 cells and wing discs at concentrations found in zeocin (Invitrogen), and 400 μg/mL G418 (Invitrogen). hemolymph/human serum. Total lipid concentration was mea- HEK293S-TetR cells, which stably express an inducible SMO sured according to ref. 11. construct, were described (2). Cells were maintained in medium containing DMEM/F-12, 10% (vol/vol) FCS, 2 mg/mL Ge- Preparation of Saponification-Resistant Lipids. Dried VLDL lipid neticin, and 5 μg/mL Blasticidin, as described. extracts were incubated at 80 °C with 2 mL of 0.3 M methanolic NIH 3T3/Smo-mEos2 cells, which are NIH 3T3 cells that stably potassium hydroxide for 1 h. After cooling down to room tem- express Smo-mEos2 construct, were described (3). Cells were perature, the solution was extracted by two washes with diethyl maintained in DMEM + 10% (vol/vol) FCS. ether. The ether fractions, which contain saponification-resistant lipids, were combined and subjected to further analysis. Fly Stocks. Oregon R flies, hs-flippase, and adh-GAL4 obtained from Bloomington Stock Center, transgenic line UAS < HcRed > Preparation of Shh Applied in the ShhLIGHT2 Cell Assay. To prepare dsLpp is described in ref. 4. nonlipoprotein-associated Shh, we transfected HeLa cells with cDNA encoding human Shh (OriGene; SC300021) by using Lipid Extraction from Drosophila Larval Hemolymph. Third instar polyethylenimine (Polysciences) in OptiMEM (Invitrogen), then larvae were washed in 10% (vol/vol) NaCl and disrupted by using switched Shh-transfected HeLa cells to serum-free media + a loose dounce homogenizer, which releases the hemolymph but (DMEM 1% Insulin-Transferrin- mixture (ITS-X, is not designed to disrupt the cells, in 1% acetic acid in H O with Gibco) 4 h after transfection. After 48 h, conditioned media were 2 × 50 μM PF-3845. The extract was centrifuged at 1,000 × g for centrifuged at 1,000 g for 20 min and concentrated by using 10 min. The supernatant containing hemolymph was centrifuged Amicon Ultra -10K (Millipore) for use in the signaling assay. at 16,000 × g for 2 h to remove the debris and subjected to lipid Identically treated media from nontransfected HeLa cells extraction. were used as controls. To prepare Lipoprotein-Shh, transfected HeLa cells were cultured in DMEM + 10% (vol/vol) FBS. Con- Reagents. Lipids used in this study were as follows: cholesterol, ditioned media were centrifuged at 1,000 × g for 20 min. For desmosterol, 7-dehydrocholesterol, vitamin D3 (Avanti Polar signaling ShhLIGHT2 assay, lipoprotein-Shh was isolated by Lipids), ergosterol, ergocalciferol D2, 1,25-dihydroxyvitamin D3 density centrifugation (12) and concentrated by using Amicon (Sigma-Aldrich), N-acylethanolamides 16:0, 16:1, 18:0, 18:1, Ultra -10K. 18:2, 20:4, N-acyldopamine 16:0, N-acyldopamine 20:4, 2-alkyl- ShhLIGHT2 Signaling and SAG Competition Assays. Twenty-four glycerol 18:2, 2-acylglycerol 20:4, 2-alkylglycerol 20:4 and 5 N-acylserine 20:4 (Cayman Chemical), N-acyl dopamine 18:0 hours before assay, ShhLIGHT2 cells were plated at 10 per well (Tocris Bioscience), N-acyl serine 16:0 (Avanti Polar Lipids), in 96-well plates and then switched to a serum-free medium + delta-9- (Biomol), and consisting of DMEM 1% ITS-X. In this medium, cells were (Sigma-Aldrich), ACEA, AM251, AM404, AM630, CB-13, O-1918, then treated with nontransfected serum-free HeLa cell super- CP-775146, GW6471, PF-3845, S 26948 and T 0070907 (Tocris natants and DMSO or DOPC liposomes (mock) or with non- Bioscience), SMO Agonist (SAG) (Santa Cruz Biotechnology), and lipoprotein-associated Shh supplemented with DMSO or DOPC BODIPY-cyclopamine (BioVision). Deuterated endocannabinoids liposomes (Shh) or with different lipids or synthetic compounds. used for LC-MS/MS quantification of endogenous species were Purified, commercially obtained lipids, or synthetic compounds purchased from Biomol and R&D Systems. were added in DMSO at indicated concentration. HPLC elution fractions were added in DOPC liposomes in Induction of RNAi. LppRNAi was induced as described (4). a ratio of 1:4. SAG was added to the ShhLIGHT2 cells at indicated concen- Immunohistochemistry and Image Analysis. Imaginal discs were trations alone or in the presence of different endocannabinoids. fixed and stained as described (5). Image analysis and statistics of Luciferase activity was assayed in cell lysates after 24 h, as image quantifications were performed as described (6). Anti- instructed by the manufacturer (Dual Glo Luciferase Assay; bodies were diluted as follows: anti-Ci 2A1 1:10 (7) and anti- Promega). The resulting Hh pathway activity was measured as the SMO 1:50 (8). firefly:Renilla ratio normalized to the ratio in cells that were not stimulated with Shh. Lipoprotein Isolation. VLDL, LDL, and HDL were isolated from human serum (Sigma) according to ref. 9. Drosophila Lipophorin BODIPY-Cyclopamine Binding Assay. HEK293S-TetR cells were (Lpp) was isolated as described (6). grown to confluence in medium containing Geneticin (2 mg/mL). The growth medium was then replaced with fresh medium Lipid Extraction from Drosophila Larval Hemolymph. Third instar containing tetracycline (2 μg/mL) and sodium butyrate (5 mM). larvae were washed in 10% (vol/vol) NaCl and disrupted by using After 2 d, fluorescence binding assays using BODIPY-cyclop- a loose dounce homogenizer, which releases the hemolymph but amine were conducted as described (13). BODIPY fluores- is not designed to disrupt the cells, in 1% acetic acid in H2O with cence was monitored by FACS and analyzed by using the 50 μM PF-3845. The extract was centrifuged at 1,000 × g for FlowJo software.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 1of9 Ciliary SMO Localization Assay. NIH 3T3/Smo-mEos2 cells were normal goat serum), incubated with the anti-acetylated Tubulin grown on glass coverslips to reach confluence. The cells were then antibody (Sigma, 1:2,000) in blocking solution for 1 h, washed with shifted to DMEM + 1% ITS-X and incubated for 24 h with or PBS, incubated with the secondary antibody in blocking solution, without SAG and different compounds. Cells were fixed for washed, and mounted in Vectashield. Slides were viewed under 10 min with 4% (vol/vol) in PBS, incubated for the confocal microscope Zeiss LSM Meta and analyzed by using 20 min in blocking solution (PBS + 0.1% Triton X-100 + 1% the Fiji software.

1. Taipale J, Cooper MK, Maiti T, Beachy PA (2002) Patched acts catalytically to suppress 8. Lum L, et al. (2003) Hedgehog signal transduction via Smoothened association with the activity of Smoothened. Nature 418(6900):892–897. a cytoplasmic complex scaffolded by the atypical kinesin, Costal-2. Mol Cell 12(5): 2. Dwyer JR, et al. (2007) Oxysterols are novel activators of the hedgehog signaling 1261–1274. pathway in pluripotent mesenchymal cells. J Biol Chem 282(12):8959–8968. 9. Redgrave TG, Roberts DC, West CE (1975) Separation of plasma lipoproteins by 3. Kim J, et al. (2014) Simultaneous measurement of smoothened entry into and exit density-gradient ultracentrifugation. Anal Biochem 65(1-2):42–49. from the primary cilium. PLoS ONE 9(8):e104070. 10. Bligh EG, Dyer WJ (1959) A rapid method of total lipid extraction and purification. 4. Panáková D, Sprong H, Marois E, Thiele C, Eaton S (2005) Lipoprotein particles are Can J Biochem Physiol 37(8):911–917. required for Hedgehog and Wingless signalling. Nature 435(7038):58–65. 11. Marsh JB, Weinstein DB (1966) Simple charring method for determination of lipids. J 5. Marois E, Mahmoud A, Eaton S (2006) The endocytic pathway and formation of the Lipid Res 7(4):574–576. Wingless morphogen gradient. Development 133(2):307–317. 12. Rudel LL, Lee JA, Morris MD, Felts JM (1974) Characterization of plasma lipoproteins 6. Khaliullina H, et al. (2009) Patched regulates Smoothened trafficking using lipopro- separated and purified by agarose-column chromatography. Biochem J 139(1):89–95. tein-derived lipids. Development 136(24):4111–4121. 13. Chen JK, Taipale J, Cooper MK, Beachy PA (2002) Inhibition of Hedgehog signaling by 7. Wang QT, Holmgren RA (1999) The subcellular localization and activity of Drosophila direct binding of cyclopamine to Smoothened. Genes Dev 16(21):2743–2748. cubitus interruptus are regulated at multiple levels. Development 126(22):5097–5106.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 2of9 Fig. S1. Lipids derived from mammalian lipoproteins reduce SMO levels in Drosophila wing imaginal discs and inhibit Shh signaling in ShhLIGHT2 cells. (A) Ratio of firefly:Renilla luciferase activity in ShhLIGHT2 cells treated as indicated. Treatment with DMSO served as a control for background activity (-). Error bars indicate SDs of three independent experiments. n > 3. (B′–B****) Wing discs from wild-type (B′) and LppRNAi (B*–B****) animals mock-treated (B′ and B*) or treated with HDL- (B**), LDL- (B***), or VLDL-derived (B****) lipids. (C) Quantification of SMO staining in the wing disk assay performed in (B′–B****) with lipids derived from HDL (magenta; P = 1.4), LDL (blue; P = 0.0048 for far anterior) or VLDL (green; P = 1.0 × 10−11). Anterior is to the left; n > 10 discs for each quantification. (D) Wing discs from wild type (D′ and D*) and LppRNAi (D′′ and D**) animals mock-treated (D′ and D′′) or with nonsaponifiable Lpp lipids (D* − and D**; P = 1.4 × 10 12) and stained for SMO. Anterior is to the left. (E) Quantification of SMO staining in the wing disk assay performed in (D′–D**). P = 1.4 × − 10 12 for LppRNAi wing discs treated with nonsaponifiable Lpp lipids. n >10 discs for each quantification. (F) Ratio of reporter activity in ShhLIGHT2 cells treated with Shh or with Shh and nonsaponifiable Lpp lipids. Treatment with DMSO served as a control for background activity (-). Error bars indicate SDs of five independent experiments. n = 5. (G) Wing discs from wild type (G’ and G*) and LppRNAi (G’’ and G**) animals mock-treated (G’ and G’’) or with non- saponifiable VLDL lipids (G* and G**; P = 4.1 × 10−33) and stained for SMO. Anterior is to the left. (H) Quantification of SMO staining in the wing disk assay performed in (D′–D**). P = 4.1 × 10−33 for LppRNAi wing discs treated with nonsaponifiable VLDL lipids. n >10 discs for each quantification. (I) Ratio of reporter activity in ShhLIGHT2 cells treated with Shh or with Shh and nonsaponifiable Lpp lipids. Treatment with DMSO served as a control for background activity (-). Error bars indicate SDs of five independent experiments. n = 5. AP, anteroposterior. (Scale bars: 10 μm.)

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 3of9 Fig. S2. Sphingolipids and 20-hydroxycholesterol do not cofractionate with the inhibitory or stimulatory activities present in VLDL. (A) Ratio of reporter activity in ShhLIGHT2 cells treated as indicated. Error bars indicate SDs of three separate measurements of fraction activity. n = 3. (B–F) Distribution of different sphingolipid species (indicated) in elution fractions tested in A.(G) Distribution of 20-hydroxycholesterol in elution fractions tested in A. List of acquisitions and queries used for identification of the different compounds can be found in Table S1.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 4of9 Fig. S3. Specific activities of endocannabinoids and phytocannabinoids toward Hedgehog signaling and receptors 1 and 2 (CB1 and CB2), PPARα, PPARβ/δ, PPARγ, GPR55, GPR119, TRP channels, and fatty acid amide hydrolase (FAAH). Values in the columns are EC50 values (micromolars) for the signaling pathway induced by the indicated . References for the published values are indicated as superscripts.

1. Rhee MH, et al. (1997) Cannabinol derivatives: Binding to cannabinoid receptors and inhibition of adenylylcyclase. J Med Chem 40(20):3228–3233. 2. Pertwee RG (2008) The diverse CB1 and CB2 receptor pharmacology of three plant : Delta9-tetrahydrocannabinol, cannabidiol and delta9-. Br J Pharmacol 153(2):199–215. 3. Gonsiorek W, et al. (2000) Endocannabinoid 2-arachidonyl glycerol is a full agonist through human type 2 : Antagonism by . Mol Pharmacol 57(5): 1045–1050. 4. Cohen-Yeshurun A, et al. (2011) N-arachidonoyl-L-serine is neuroprotective after traumatic brain injury by reducing apoptosis. J Cereb Blood Flow Metab 31(8):1768–1777. 5. Milman G, et al. (2006) N-arachidonoyl L-serine, an endocannabinoid-like brain constituent with vasodilatory properties. Proc Natl Acad Sci USA 103(7):2428–2433. 6. Song ZH, Slowey CA, Hurst DP, Reggio PH (1999) The difference between the CB(1) and CB(2) cannabinoid receptors at position 5.46 is crucial for the selectivity of WIN55212-2 for CB(2). Mol Pharmacol 56(4):834–840. 7. Hanus L, et al. (2001) 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci USA 98(7):3662–3665. 8. Chu CJ, et al. (2003) N-oleoyldopamine, a novel endogenous -like lipid that produces hyperalgesia. J Biol Chem 278(16):13633–13639. 9. Bisogno T, et al. (2000) N-acyl-dopamines: Novel synthetic CB(1) cannabinoid-receptor ligands and inhibitors of anandamide inactivation with cannabimimetic activity in vitro and in vivo. Biochem J 351(Pt 3):817–824. 10. O’Sullivan SE (2007) Cannabinoids go nuclear: Evidence for activation of peroxisome proliferator-activated receptors. Br J Pharmacol 152(5):576–582. 11. Shoemaker JL, Joseph BK, Ruckle MB, Mayeux PR, Prather PL (2005) The endocannabinoid noladin ether acts as a full agonist at human CB2 cannabinoid receptors. J Pharmacol Exp Ther 314(2):868–875. 12. LoVerme J, La Rana G, Russo R, Calignano A, Piomelli D (2005) The search for the receptor. Life Sci 77(14):1685–1698. 13. Sun Y, Alexander SP, Kendall DA, Bennett AJ (2006) Cannabinoids and PPARalpha signalling. Biochem Soc Trans 34(Pt 6):1095–1097. 14. Kozak KR, et al. (2002) 15-Lipoxygenase metabolism of 2-arachidonylglycerol. Generation of a peroxisome proliferator-activated receptor alpha agonist. J Biol Chem 277(26):23278– 23286. 15. Sun Y, Bennett A (2007) Cannabinoids: A new group of agonists of PPARs. PPAR Res 2007:23513. 16. Fu J, et al. (2003) Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-alpha. Nature 425(6953):90–93. 17. Ghosh M, et al. (2007) COX-2 suppresses tissue factor expression via endocannabinoid-directed PPARdelta activation. J Exp Med 204(9):2053–2061. 18. O’Sullivan SE, Tarling EJ, Bennett AJ, Kendall DA, Randall MD (2005) Novel time-dependent vascular actions of Delta9-tetrahydrocannabinol mediated by peroxisome proliferator- activated receptor gamma. Biochem Biophys Res Commun 337(3):824–831. 19. Bouaboula M, et al. (2005) Anandamide induced PPARgamma transcriptional activation and 3T3-L1 preadipocyte differentiation. Eur J Pharmacol 517(3):174–181. 20. Rockwell CE, Snider NT, Thompson JT, Vanden Heuvel JP, Kaminski NE (2006) Interleukin-2 suppression by 2-arachidonyl glycerol is mediated through peroxisome proliferator- activated receptor gamma independently of cannabinoid receptors 1 and 2. Mol Pharmacol 70(1):101–111. 21. LoVerme J, et al. (2006) Rapid broad-spectrum analgesia through activation of peroxisome proliferator-activated receptor-alpha. J Pharmacol Exp Ther 319(3):1051–1061. 22. Lauckner JE, et al. (2008) GPR55 is a cannabinoid receptor that increases intracellular and inhibits M current. Proc Natl Acad Sci USA 105(7):2699–2704. 23. Waldeck-Weiermair M, et al. (2008) Integrin clustering enables anandamide-induced Ca2+ signaling in endothelial cells via GPR55 by protection against CB1-receptor-triggered repression. J Cell Sci 121(Pt 10):1704–1717. 24. Ryberg E, et al. (2007) The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol 152(7):1092–1101. 25. Chu ZL, et al. (2010) N-oleoyldopamine enhances homeostasis through the activation of GPR119. Mol Endocrinol 24(1):161–170. 26. Hansen KB, et al. (2011) 2-Oleoyl glycerol is a GPR119 agonist and signals GLP-1 release in humans. J Clin Endocrinol Metab 96(9):E1409–E1417. 27. Qin N, et al. (2008) TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci 28(24):6231–6238. 28. Bisogno T, et al. (2001) Molecular targets for cannabidiol and its synthetic analogues: Effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol 134(4):845–852. 29. De Petrocellis L, Bisogno T, Davis JB, Pertwee RG, Di Marzo V (2000) Overlap between the recognition properties of the anandamide transporter and the VR1 vanilloid receptor: Inhibitors of anandamide uptake with negligible capsaicin-like activity. FEBS Lett 483(1):52–56. 30. Ahern GP (2003) Activation of TRPV1 by the satiety factor . J Biol Chem 278(33):30429–30434. 31. De Petrocellis L, et al. (2008) Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther 325(3):1007–1015. 32. Pertwee RG (2010) Receptors and channels targeted by synthetic cannabinoid receptor agonists and antagonists. Curr Med Chem 17(14):1360–1381. 33. De Petrocellis L, et al. (2007) Regulation of transient receptor potential channels of melastatin type 8 (TRPM8): Effect of cAMP, cannabinoid CB(1) receptors and endovanilloids. Exp Cell Res 313(9):1911–1920. 34. Jordt SE, et al. (2004) oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 427(6971):260–265. 35. De Petrocellis L, Di Marzo V (2009) Role of endocannabinoids and endovanilloids in Ca2+ signalling. Cell Calcium 45(6):611–624. 36. De Petrocellis L, Di Marzo V (2010) Non-CB1, non-CB2 receptors for endocannabinoids, plant cannabinoids, and synthetic cannabimimetics: Focus on G-protein-coupled receptors and transient receptor potential channels. J Neuroimmune Pharmacol 5(1):103–121. 37. Ghafouri N, et al. (2004) Inhibition of and fatty acid amide hydrolase by analogues of 2-arachidonoylglycerol. Br J Pharmacol 143(6):774–784. 38. Steffens M, Schulze-Bonhage A, Surges R, Feuerstein TJ (2005) Fatty acid amidohydrolase in human neocortex-activity in epileptic and non-epileptic brain tissue and inhibition by putative endocannabinoids. Neurosci Lett 385(1):13–17. 39. Jonsson KO, Vandevoorde S, Lambert DM, Tiger G, Fowler CJ (2001) Effects of homologues and analogues of palmitoylethanolamide upon the inactivation of the endocannabinoid anandamide. Br J Pharmacol 133(8):1263–1275.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 5of9 Fig. S4. Phytocannabinoids and N-acylethanolamide 18:2 do not prevent ciliary SMO localization. A–E show NIH 3T3/Smo-mEos2 cells not treated (A), treated with 100 nM SAG alone (B) or treated with 100 nM SAG and 1 μM Cannabidiol (C), 10 μM N-acylethanolamide 18:2 (D), or 1 μM Cannabinol (E) for 24 h. n = 2. Cells were stained for acetylated Tubulin (magenta); Smo-mEos is shown in cyan. F–J show NIH 3T3/Smo-mEos2 cells pretreated with 100 nM SAG for 24 h, followed by no treatment (F), treatment with 100 nM SAG alone (G) or treatment with 100 nM SAG and 1 μM Cannabidiol (H), 10 μM N-acylethanolamide 18:2 (I), or 1 μM Cannabinol (J)for4h.n = 2. Cells were stained for acetylated Tubulin (magenta); Smo-mEos is shown in cyan. (Scale bars: 10 μm.)

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 6of9 Fig. S5. Specific endocannabinoids and cannabinoids reduce Smo levels in Drosophila wing imaginal discs. (A–B**) Wing discs from either wild type (A’, A*, B′, and B*) or LppRNAi (A′′, A**, B′′, and B**) animals that were mock-treated (A′, A′′, B′, and B′′) or treated with 50 μM 2-acylglycerol 20:4 (A′′, A**, B′′, and B**), stained for Smo (A′–A**) and Ci155 (B′–B**). A and B show quantification of Smo (A) and Ci155 (B) staining in the corresponding discs. Anterior is to the left; AP, − − − anteroposterior. (Scale bars: 10 μm.) n > 10 discs for each quantification (P = 1.3 × 10 18 for anterior, P = 3.8 × 10 13 for posterior in wild-type in A; P = 2.8 × 10 34 − for anterior, P = 1.0 × 10 20 for posterior in LppRNAi in B). (C and D) Quantifications of Smo staining in wing disk assays performed with 50 μM N-acylethanolamide 18:1 (pink; P = 0.41) (C)and50μM N-acylethanolamide 18:0 (green; P = 0.74) or 50 μM N-acylethanolamide 20:4 (blue; P = 1.6) (D). (C) Quantification of Smo

Legend continued on following page

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 7of9 staining in wing disk assay performed with 50 μM N-acylethanolamide 18:2 (red; P = 1.2 × 10−6), 50 μM 2-acylglycerol 16:0 (brown; P = 1.3 × 10−5), 50 μM − Cannabidiol (violet; P = 0.00109), and 50 μM Cannabinol (blue; P = 4.3 × 10 14). Anterior is to the left; AP, anteroposterior. n > 10 discs for each quantification.

Fig. S6. Effects of vitamin D3 on Shh signaling in ShhLIGHT2 cells or SMO stability in Drosophila wing imaginal discs. Effect of N-acylserine 16:0 on ShhLIGHT2 cells. (A–C) Effects of increasing concentrations of 7-dehydrocholesterol (A), vitamin D3 (B), and Dihydroxyvitamin D3 (C) on signaling by nonlipoprotein- associated Shh in ShhLIGHT2 cells. Ratio of background firefly:Renilla luciferase activity (treatment with DMSO alone) is defined as 0. Maximum ratio of re- porter activity (ranged between six- and eightfold over background) in cells treated with nonlipoprotein-associated Shh alone is normalized to 100 and is indicated by green line. Error bars indicate SDs of at least five independent experiments. n = 5. (D) Quantification of SMO staining in wild-type (gray) and LppRNAi wing discs that have been mock-treated (orange) or treated with 100 μM vitamin D3 (pink; P = 0.44). Anterior is to the left; AP, anteroposterior. n > 10 discs for each quantification. (E) Effects of increasing concentrations of N-acylserine 16:0 on firefly/Renilla luciferase activity in ShhLIGHT2 cells. Red line shows the firefly/Renilla luciferase activity in cells treated with Shh alone, and the firefly/Renilla luciferase activity in not-treated cells is indicated by green line. Error bars indicate SDs of at least three independent experiments. n = 2.

Table S1. List of acquisitions and queries used for identification of the different compounds Detection mode Elemental composition Fragment ion mass Precursor ion mass Lipid class Polarity (fragment mass(es)) of precursor ions (error in mmu) (error in mmu)

+ N-acylethanolamides + MS/MS (62.0600) Cx Hx NO2 (H ) 62.0608 (+0.73) — + 2-alkylglycerols + MS/MS (75.0441) Cx Hx NO3 (NH4 ) 75.0449 (+0.83) — + N-acyldopamines + MS/MS (154.0863) Cx Hx NO3 (H ) 154.0862 (−0.10) — + N-acylserines — MS/MS (104.0353) Cx Hx NO3 (-H ) 104.0335 (−1.8) — + 20-hydroxycholesterol + MS/MS (385.3465/367.3359) C27 H50 NO2 (NH4 ) 385.3496 (−0.18) — 367.3355 (−0.05) + 2-acylglycerols + NL (109.0739) Cx Hx O4 N(NH4 ) 109.0737 (−0.09) — + LCB-P — MS/MS (78.9590) Cx Hx O5 N P (-H ) 78.9591 (−1.4) — + LCB + FTMS (e.g., 298.2741) Cx Hx O2 N(H ) — 298.2742 (+0.15) + Ceramides + FTMS (e.g., 648.6289) Cx Hx O3–4 N(H ) — 648.6289 (−0.05) + Hexosylceramides + FTMS (e.g., 700.5722) Cx Hx O3–4 N(H ) — 700.5720 (−0.21)

Scripts used for identification and quantification can be found here (https://wiki.mpi-cbg.de/lipidx/Lx_reference_khaliullina_2015). FTMS, Fourier-transform mass spectrometry; LCB, long chain base; LCB-P, long chain base-phosphate; MS/MS, mass spectrometry; NL, neutral loss.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 8of9 Table S2. Endocannabinoids and related molecules quantified in Drosophila larval hemolymph by LC-MS/MS by the method of MRM MRM transitions

Name Precursor ion, m/z Product ion, m/z Concentration, nM

N-acylethanolamide 16:0 300.3 62.1 610.179 ± 15.78 N-acylethanolamide 18:1 326.3 62.1 2,179.999 ± 42.99 N-acylethanolamide 18:0 328.3 62.1 72.402 ± 0.39 Total N-acylethanolamides 2,862.58 N-acylserine 16:0 344.3 106.1 607.84 ± 33.73 N-acylserine 18:2 368.3 106.1 1,266.159 ± 57.86 N-acylserine 18:1 370.3 106.1 421.073 ± 14.21 N-acylserine 18:0 372.3 106.1 1,242.364 ± 40.34 Total N-acylserines 3,537.44 1-acylglycerol 16:0 331.3 239.3 1,247.673 ± 27.90 1-acylglycerol 18:2 355.3 263.3 434.783 ± 1.06 Total 1-acylglycerols 1,682.46 2-acylglycerol 16:0 331.3 239.3 211.52 ± 1.70 2-acylglycerol 18:2 355.3 263.3 687.991 ± 37.89 Total 2-acylglycerols 899.51 Total endocannabinoids 8,981.98

Concentrations correspond to the values in circulating hemolymph assuming an internal larval volume of 1 μL. The concentrations of endocannabinoids were measured in hemolymph with or without 10 μM PF-3845, which prevents endocannabinoid degradation.

Khaliullina et al. www.pnas.org/cgi/content/short/1416463112 9of9