Antibodies and chemicals
Antibodies: Anti-TTYH1 (26973-1-AP; Proteintech); anti-ALDH1L1 (NBP2-50045; Novus Biologics); anti-LAMP1 (H4A3-c; Developmental Studies Hybridoma Bank); anti- SQSTM1 (p62) (AB109012-1001; Abcam); Anti-LC3 (83506S; Cell Signaling); anti-alpha-tubulin (12G10; Developmental Studies Hybridoma Bank); anti-RFP (600-401-379; Rockland); goat anti-mouse IgG IRDye 800CW (926-32210; Li-COR); goat anti-rabbit IgG IRDye 680LT (926-68021; Li-COR); donkey anti-mouse IgG Alexa Fluor 488 (A-21202; Invitrogen); donkey anti-rabbit IgG Alexa Fluor 488 (A-21206; Invitrogen); donkey anti-mouse IgG Alexa Fluor 568 (A10037; Invitrogen); donkey anti-rabbit IgG Alexa Fluor 568 (A10042; Invitrogen).
Chemicals: Ceramide-1-phosphate (d18:1/16:0) (C1P; 22542; Cayman); bafilomycin A1 (BafA1; B1793; Sigma-Aldrich); NVP-213 (HY-13945; MedChem Express); human IL-1β (SRP3083; Sigma-Aldrich); biotinylated human IL-1β (ILB-H82E9; Acro Biosystem); bicuculine (Bic; HY-N0219; MedChem Express); 4-aminopyridine (4AP; 275875; Sigma-Aldrich); methyl tert-butyl ether (MTBE; 34875; Sigma-Aldrich).
Generation of Ttyh1flox conditional knockout mouse
The Ttyh1flox conditional knockout mouse strain was generated by the Rutgers University Genome Editing Shared Resource core. LoxP (ATAACTTCGTATAATGTATGCTATACGAAGTTAT) sites were inserted to the introns immediately flanking exon 4 of Ttyh1 in C57BL/6 strain using CRISPR/Cas9. Founder mice were cross-bred to generate homozygous Ttyh1fl/fl. Genotyping was performed by Sanger sequencing on the PCR amplicons of the loxP-inserted intron sequences using the following primers:
5’-loxP: CTGGAGACCAAAAGCAGCCAAATC and AGTTCCTGAACTTGCATCTGAGCC
3’-loxP: TGTCCCACTGGTTGAATGAGAAGC and AGACGAGTTGCACGATGAACACAG
Homozygous Ttyh1fl/fl mice were crossed with Aldh1l1-Cre/ERT2 BAC transgenic mice (031008; The Jackson Laboratory) to generate the Aldh1l1-Cre/ERT2; Ttyh1fl/fl mice. Genotyping for the Aldh1l1-Cre/ERT2 allele was performed by PCR amplification using the following primers:
CTTCAACAGGTGCCTTCCA and GGCAAACGGACAGAAGCA
Isolation and primary culture of mouse cortical neurons and astrocytes
Mouse brains of P0 newborn mice were dissected and isolated brains were placed in ice cold phosphate buffered saline (PBS; 10010023; Gibco). Meninges were removed above the cortex region and the cortex was separated, and the cortical tissues were dissected into small pieces. After washing three times with PBS, the cortical tissues were incubated with collagenase (1 mg/mL; C2674; Sigma-Aldrich) for 30 min at 37°C. Collagenase was then removed and the residual collagenase was neutralized with fetal bovine serum (FBS; F4135; Sigma-Aldrich) by incubating for 5 min at room temperature. After removing FBS, the tissues were triturated through pipette tips to make single-cell suspension. For cortical neurons, cells were suspended in neurobasal medium A supplemented with B-27 (10888022; Gibco), Glutamax (35050061; Gibco), and 1% penicillin and streptomycin. Neurons were plated on surface coated with poly-D-lysine. Medium was refreshed after 30 minutes to remove unattached cells. Neurons were maintained with 50% medium change every 2 days for 7 days before experiments. For cortical astrocytes, cells were suspended in primary astrocyte medium (A1261301; Gibco) supplemented with 1% penicillin and streptomycin, and were plated on surface coated with Geltrex (A1413201; Gibco). The culture dish was then shaken for 30 minutes on an orbital shaker before replacing medium with fresh primary astrocyte medium. Medium was refreshed every 3 days until reaching confluency for passage.
Astrocyte culture
Both primary mouse cortical astrocytes and primary human brain astrocytes (HMP202; Neuromics) were expanded and maintained in primary astrocyte medium (A1261301; Gibco) supplemented with 1% penicillin and streptomycin. For cortical astrocytes derived from Aldh1l1-Cre/ERT2; Ttyh1fl/fl mice, cells were treated with either 1 µg/mL 4-hydroxy tamoxifen (4-OHT; 14854; Cayman) or vehicle (DMSO) for one week before experiments. Astrocytes seeded for experiments were cultured in low-serum astrocyte medium (1801; ScienCell) for at least 2 days, followed by overnight serum-free medium synchronization before experiments.
Neuronal conditioned medium (NCM)
Mouse cortical neurons were seeded on 6-well plate at a density of ~50k cells per well. Mouse neurons were cultured in B-27 supplemented neural basal medium (10888022; Gibco) for 7 days before treatment and medium conditioning. For Cerk knockdown, mouse neurons were transduced with lentiviral vector and cultured for an additional 3 days. For iNeurons, cells were seeded on 24-well plate at a density of ~57k cells per well. iNeurons were cultured in STEMdiff forebrain maturation medium (08605; Stemcell Tech) for 14 days before treatment and medium conditioning. Neurons were treated with 10 ng/mL of IL-1β together with NVP-231 (12 nM; HY-13945; MedChem Express) or vehicle DMSO (0.1%) for 2 hours. IL-1β was omitted in the naïve treatment condition. After treatment, culture medium was replaced with plain neural basal medium (10888022; Gibco) for 2 hours to condition the medium. The neuronal conditioned medium (NCM) was first centrifuged for 5 minutes at 1500 g to remove cell debris. The supernatant was collected for solvent extraction of secreted lipids. After solvent extraction, dried lipids were reconstituted in 800 µL DMEM (10569-010; Gibco). Half of each lipid extract was used to treat one well of 25k astrocytes seeded in 24-well plate for 2 hours, before lysate collection for Western blotting or immunofluorescence for lipid droplets.
Neuron-astrocyte co-culture
Primary mouse cortical neurons were seeded in poly-D-lysine-coated transwell inserts (230635; CELLTREAT) and cultured in B-27 supplemented neurobasal medium for 7 days. For shRNA-mediated Cerk knockdown, neurons were transduced with lentiviral vectors and cultured for an additional 3 days. One day before coculture, mouse cortical astrocytes were seeded in 24-well plates (P24-1.5P; Cellvis) and cultured in neurobasal medium. The neuron-containing transwells were then inserted into the astrocytes-seeded wells. Neurons and astrocytes were co-cultured in neurobasal medium for 1 day before addition of beads to the transwell. Approximately 1.9 nmol of biotinylated human IL-1β (ILB-H82E9; Acro Biosystem) was incubated with 0.5 mg of streptavidin magnetic beads (CCT-1497; Vector Laboratories) in DMEM/F-12 medium (12660012; Gibco) for 30 minutes before washing with DMEM/F-12 for 3 times on a magnetic rack. The IL-1β-conjugated beads were resuspended at 1 µg IL-1β (12.5 mg beads) per milliliter of DMEM/F-12 (100x stock). Control unconjugated beads were prepared the same way omitting IL-1β. Beads were added at 1/100 dilution to the neuron-containing transwells for additional 24 hours of co-culture. Astrocytes in the bottom wells were then either stained for lipid droplets or lysed for lipid extraction.
Differentiation of human neurons (iNeurons) and astrocytes (iAstrocytes) from iPSC-derived neural progenitor cells (NPCs)
Human iPSC-Derived NPCs were differentiated from the iPSC line SCTi003-A and were obtained directly from manufacturer Stemcell Technologies (200-0620; Stemcell Tech). The NPCs were cultured in STEMdiff Neural Progenitor Medium (05833; Stemcell Tech) for fewer than 4 passages before differentiation into iNeurons and iAstrocytes, respectively. Neuronal differentiation was performed using the STEMdiff Forebrain Neuron Differentiation kit (08600; Stemcell Tech). NPCs were seeded on culture plate coated with poly-L-ornithine and laminin (PLO+Lam), and were cultured in STEMdiff Forebrain Neuron Differentiation Medium (08600; Stemcell Tech) for one week. Cells were then passaged and seeded on 24-well plate coated with PLO+Lam and cultured in STEMdiff Forebrain Neuron Maturation Medium (08605; Stemcell Tech) for two to three weeks. The resulting iNeurons were validated by the expression of MAP2 and β-III-tubulin detected by Western blot of cell lysate and immunofluorescence of fixed cells. For iAstrocytes, differentiation was performed by adapting the protocol developed by TCW et al 1. NPCs were seeded on culture plate coated with Geltrex (A1413202; Gibco) and cultured in astrocyte medium (1801, ScienCell). Cells were passaged when confluency reached 80% and maintained for at least three weeks, before assessing for astrocytic markers and transduction with shRNA-containing lentiviral vectors. Expression of astrocytic markers, GFAP and ALDH1L1, was validated by Western blot of cell lysate. After transduction with lentiviral vectors, iAstrocytes were cultured in astrocyte medium containing 10 µg/mL hygromycin for at least two weeks. The resulting shRNA-expressing iAstrocytes were then cultured in serum-free astrocyte medium for at least three days before passage and seeding for experimentations.
shRNA-mediated knockdown
Lentiviral vectors carrying shRNAs targeting human TTYH1 and mouse Cerk were synthesized by GeneCopoeia. For human TTYH1 knockdown in iAstrocytes, the psi-LVRU6MH vector containing U6 promoter, mCherry expression construct, and hygromycin resistance cassette was used. The target sequences for TTYH1 and control shRNAs are as follows.
TTYH1: GGCATTGGCATCGGTTTCTAT
Control: GCTTCGCGCCGTAGTCTTA
For Cerk knockdown in mouse cortical neurons, the psi-lvru6gp vector containing U6 promoter, GFP expression construct, and puromycin resistance cassette was used. The target sequences for Cerk and control shRNAs are as follows.
Cerk: AATCCGTTCGCATTCACAGTC
Control: GCTTCGCGCCGTAGTCTTA
Lentiviral vectors were produced using the 3rd generation packaging system and further purified by centrifugation in lentivirus concentrator (Viral-PEG LVG100; OZBioscience). Upon transduction, iAstrocytes were cultured with 10 µg/mL hygromycin for at least two passages. Hygromycin was withdrawn when cells were seeded for experiments. For mouse cortical neurons, cells were not subject to selection and were used for experiments three days after transduction.
Generation of expression constructs
Complementary DNA sequence of C-terminally RFP-tagged human TTYH1 (NM_001005367.2) was synthesized as described previously 2. Site-direct mutagenesis to generate the TTYH1ER/AA mutant was performed using NEBuilder HiFi (New England Biolabs) and the following primers (mutated codons are underlined): TGGCGGAGTACGCGTGGCTGGCCTA and CACGCGTACTCCGCCACAAAGGACACA. The cDNA sequences of TTYH1 and TTYH1ER/AA were respectively subcloned into expression vector pQCXI-Puro, which drives transgene expression by a CMV promoter.
Transfection of astrocytes with expression constructs
Astrocytes were transfected with expression constructs of GFP-LC3-RFP-LC3ΔG, TTYH1, or TTYH1ER/AA using Lipofectamine 3000 (L3000001; Invitrogen). Plasmid DNA of pMRX-IP-GFP-LC3-RFP-LC3ΔG was a kind gift from Dr. Noboru Mizushima and was obtained from Addgene. Four hours after transfection, transfection complex in Opti-MEM reduced serum medium (31985070; Invitrogen) was removed and replaced by fresh astrocyte medium. One day after transfection, cells were synchronized in serum-free medium overnight, before recovery in astrocyte medium for 4 hours. Cells were then treated before imaging or lysate collection for Western blotting.
Western blotting
Cells were lysed in RIPA buffer (9806S; Cell Signaling) supplemented with protease and phosphatase inhibitor cocktail (78447; Thermo Scientific). Laemmli buffer (161-0747; Bio-Rad) was added to lysate before loading onto 4-20% gradient gel (4561096; Bio-Rad) for electrophoresis. Separated proteins were transferred onto nitrocellulose membrane (10600004; Amersham), followed by blocking (927-80001; Li-COR). Membrane was cut at the 25 kDa ladder mark into two parts. The lower part was used for LC3 detection while the upper part was used for other protein targets. After incubation with primary antibodies, IRDye 680LT- and IRDye 800CW-conjugated secondary antibodies were used to detect the protein targets on the membrane. Membrane image captured by a ChemiDoc MP imager (BioRad) was exported to TIFF image file. The images were processed and analysed using ImageJ (National Institutes of Health).
Immunofluorescence
Cultured cells: Astrocytes seeded on glass-bottom dishes were cultured in astrocyte medium (1801; ScienCell) for 48 hours before overnight synchronization in serum-free medium. Cells were fixed with 4% paraformaldehyde in PBS for 30 minutes at room temperature. After washing three times with PBS, cells were permeabilized by incubating with 0.1% Triton X-100-containing PBS (PBST) for 15 minutes. Samples were then blocked with 5% normal donkey serum (017-000-121; Jackson Immuno). Samples were incubated overnight at 4℃ in primary antibodies diluted in PBST with 1% BSA (PBST-BSA). After primary antibody incubation, samples were washed three times with PBS and incubated with the fluorophore-conjugated secondary antibodies in PBST-BSA for one hour at room temperature in the dark. After washing with PBST for three times, samples were immersed in mounting medium with DAPI (P36985; Invitrogen). Images were taken using confocal microscopy (Zeiss LSM 980 with Airyscan; 100x oil objective) with appropriate laser power and filter selection. Representative images were processed and generated using Zen 3.3 (Zeiss) and ImageJ (National Institutes of Health).
Mouse brain tissues: Mice were fully anesthetized with a cocktail of ketamine (K; 100 mg/kg), xylazine (X; 10 mg/kg), and acepromazine (A; 10 mg/kg) (KXA; 0.1 ml/10 g body weight; intraperitoneal injections). They were then perfused transcardially through the left ventricle with cold PBS followed by 4% paraformaldehyde (PFA) in PBS. The brains were placed in 30% sucrose after 24-hour fixation in PFA. Brains were sectioned and stored in an antifreeze solution at −20°C. Coronal sections (40 μm thick) spanning the entire brain from anterior to posterior were prepared in serial order. For histological analysis, an antigen retrieval procedure was performed as previously described 3. Briefly, the sections were placed in hot citrate buffer (1.8 mM citric acid, 8.2 mM tri-sodium citrate) and incubated for 7 minutes. The sections were then cooled to room temperature in the citrate buffer for 1 hour. Immunostaining was conducted with primary antibodies, followed by secondary antibodies. DAPI (D1306; Molecular Probes) was used for counterstaining. Images were acquired using a Leica THUNDER Imaging System with a multi-track configuration.
Lipid droplet staining
Astrocytes were synchronized with serum free medium supplemented with palmitic acid (PA) conjugated BSA (50 µM PA and 142 µM BSA) for overnight before experiments. After respective treatments, cells were stained with 10 µM BODIPY493/503 (D3922; Invitrogen) for 30 minutes at room temperature. After washing with PBS, cells were fixed with 4% paraformaldehyde for 30 minutes. Fixed cells were washed with PBS for three times before mounting with mounting medium containing DAPI. Images taken by confocal microscopy were processed by CellProfiler (Broad Institute) for analyses and by ImageJ for representative images.
NBD-C1P pulse-chase assay
Ttyh1 WT and Ttyh1 KO astrocytes were seeded on Geltrex-coated glass-bottom culture dish and cultured in astrocyte medium (1801; ScienCell) for one to two days. Cells were then incubated with 2 µM NBD-C1P (S-500N6, Echelon Biosciences) in 0.005% fatty acid free bovine serum albumin (A7030; Sigma-Aldrich) supplemented artificial cerebrospinal fluid (ACSF; 126 mM NaCl, 24 mM NaHCO3, 0.375 mM KCl, 1.25 mM KH2PO4, 2 mM MgCl2, 1 mM CaCl2, 1 mM glucose; pH 7.4) for 15 minutes at 37 °C. After washing with ACSF twice to get rid of extracellular NBD-C1P, cells were either fixed with 4% paraformaldehyde (0-min chase) or chased in ACSF with 0.5 µM LysoTracker (L7528; Invitrogen) at 37 °C for 15, 30, or 60 minutes. Chase was terminated by fixation in 4% paraformaldehyde for 15 minutes. Fixed cells were immersed in DAPI Floromount-G mounting medium (Southern Biotech) before imaging. NBD-C1P fluorescence intensity was measured by imaging single cells using a Zeiss Axio Observer microscope equipped with a Colibri 5 LED light source and a 90 HE LED multi-bandpass filter with excitation wavelength/bandwidth of 469/38 nm. Ttyh1 WT and KO samples corresponded to the same chasing time-point were imaged successively within 1 hour. Images were captured using the same exposure ang image export settings for each WT and KO pair. Some of the samples were imaged again using a Zeiss LSM 980 with Airyscan2 confocal microscope to generate high-resolution subcellular localization images.
Image analyses
Lipid droplets: A CellProfiler (Broad Institute) pipeline was built for lipid droplet analysis. Confocal images were loaded into the pipeline as merged channel images (405 nm laser channel and 488 nm laser channel). The 405 nm laser channel image was used to detect cell nuclei, while the 488 nm laser channel image was optimized to highlight primary objects (lipid droplets) after being enhanced. The nuclei signal was utilized to generate a mask on the 488nm laser channel image to exclude lipid droplets within nuclei. The 488 nm laser channel image was also used to delineate the cell area by thresholding the signals. Cytoplasmic lipid droplet signals within the cell boundary were counted by the pipeline. Both lipid droplet and cell areas were computed for quantification.
GFP-LC3-RFP-LC3ΔG: ImageJ (National Institutes of Health) was employed to measure the fluorescence intensity of GFP-LC3-RFP-LC3ΔG. Images of GFP and RFP fluorescence emissions were captured by a Zeiss Axio Observer epifluorescence microscope with a 40x oil objective. A cell boundary mask was generated by thresholding the GFP- and RFP-channel images. The mask was used for cell area measurement. GFP and RFP emission intensities within the respective masks were then measured. Mean intensity was generated by standardizing total intensity within mask to cell area. Ratio between the mean GFP intensity and the mean RFP intensity for each cell was then calculated.
NBD-C1P pulse-chase analysis: A CellProfiler (Broad Institute) pipeline was built for evaluating the NBD fluorescence intensity across different time points (0 minutes, 15 minutes, 30 minutes, 60 minutes). Images were captured by a Zeiss Axio Observer epifluorescence microscope with a 100x oil objective. A cell boundary mask was generated by thresholding. The mask was used for cell area measurement. NBD emission intensity within the mask was then measured. Mean intensity was generated by standardizing total intensity within mask to cell area.
Fly strains and husbandry
Flies were grown with standard fly food (3 L of food contained 25.6 g agar, 80.6 g brewer’s yeast, 190.6 g of cornmeal, 40.3 g of sugar, 16.6 g of propionic acid, and 13.3 g of tegosept) in a 25°C-incubator with 12-hour light-dark cycle. The wild-type (WT; w1118) and tty-/- (ttyMI12269) strains were described previously 2. The following fly strains were obtained from the Bloomington Drosophila Stock Center: repo-GAL4 7415, UAS-nucRFP 30556, nSyb-GS 80699 (neuron-switch), UAS-Cerk 38410, UAS-Cerk-RNAi 67256 (Cerk-IR; TRiP.HMC06359). To induce neuronal expression, 1-week-old flies were fed with standard fly food containing 500 µM RU486 (mifepristone; 459980010, Acros Organics) for 2 weeks. After feeding with either regular or RU486-containing fly food, fly heads of 3-week-old flies were dissected and collected for solvent extraction of brain lipids.
Primary culture and lipid droplet staining of fly brain cells
The procedures for primary fly brain cell culture were described previously 2,4. Briefly, wandering third-instar larvae were washed with 70% ethanol and sterile water successively. Brains were dissected from pinned larvae submersed in Schneider’s medium (Sigma-Aldrich) supplemented with 10% heat-inactivated FBS and 1% antibiotic/antimycotic solution (Sigma-Aldrich). Dissected brains were transferred to filtered HL-3 solution (70 mM NaCl, 5 mM KCl, 1 mM CaCl2, 20 mM MgCl2, 10 mM NaHCO3, 115 mM sucrose, 5 mM trehalose, and 5 mM HEPES) containing 0.423 mM L-cysteine (Sigma-Aldrich) and 5 U/mL papain (Worthington) and incubated at ~29 for 20 minutes. The brains were washed twice with Schneider’s medium before triturating the tissues through the pore of a 300 µL pipette tip in supplemented Schneider’s medium. Dissociated cells were seeded in 35 mm glass-bottom dishes (Cellvis) precoated with concanavalin A (Sigma-Aldrich) and cultured in supplemented Schneider’s medium at 25 °C for 4 days.
For lipid droplet fluorescence imaging, primary glia expressing nuclear RFP driven by repo-GAL4 were incubated with 10 µM BODIPY493/503 (D3922; Invitrogen) in either HL-3 or sugar-free HL-3 (sucrose and trehalose replaced by 120 mM mannitol) for ~15 minutes before imaging using a Zeiss Axio Observer microscope equipped with a Colibri 5 LED light source and a 90 HE LED multi-bandpass filter with excitation wavelength/bandwidth of 469/38 nm and 555/30 nm. The recorded signals of sequentially fluorescence emission from Em469ex and Em555ex were analysed by CellProfiler (Broad Institute), where Em555ex signals were selected to identify locations of glia and Em469ex signals were selected to identify punctate structures representing lipid droplets. The total area of lipid droplet per cell was then measured by a CellProfiler pipeline. For each genotype and treatment group, the average lipid droplet area of a total of 30 individual cells randomly selected were compared, using the average value of wildtype glia in HL-3 as the baseline.
Lipid extraction
Lipids were extracted from cells, fly heads and conditioned medium using organic methyl tert-butyl ether (MTBE) solvent phase separation. Cells or fly heads were homogenized in standard diluent assay buffer (700732; Cayman) at 100 µL per 1 million cells or 2 µL per fly head, respectively. MTBE (34875; Sigma-Aldrich) was added to the lysate/conditioned medium at 2:1 volume ratio. The mixture was vortexed for 30 seconds. Upon 10-minute incubation at room temperature, the mixture was centrifuged at 1,000 g for 10 minutes. The upper (organic) phase was collected which contains the extracted lipids. The separated organic phase was dried in a vacuum centrifuge. Dry lipid extracts were stored at -80°C.
Lipidomics
Total lipids were extracted from adult Drosophila heads using the MTBE solvent extraction method. For each biological replicate, a total of 40 fly heads collected from snap freezing was homogenized in MTBE at 4°C. After the addition of methanol spiked with SPLASH LIPIDOMIX (330707; Avanti) as the internal standard, the mixture was mixed briefly and incubated for 5 minutes. At endpoint, the upper organic phase was transferred into a collection tube and dried. LC-MS lipid profiling was performed by the Rutgers University Metabolomics Shared Resource core. Reverse-phase separation of the lipid extract was conducted on a Vanquish Horizon UHPLC system (Thermo Fisher Scientific) using a Poroshell 120 EC-C18 column (2.7 μm particle size, Agilent InfinityLab). The gradient elution employed solvent A (90% H2O:10% MeOH with 34.2 mM acetic acid, 1 mM ammonium acetate, pH 9.4) and solvent B (75% IPA:25% methanol with 34.2 mM acetic acid, 1 mM ammonium acetate, pH 9.4). The gradient program was set as follows: 0 min, 25% B; 2 min, 25% B; 5.5 min, 65% B; 12.5 min, 100% B; 19.5 min, 100% B; 20.0 min, 25% B; 30 min, 25% B. The flow rate was set to 200 μL/min, with an injection volume of 5 μL, and the column temperature maintained at 55 °C. The autosampler was kept at 4 °C. Full-scan mass spectrometry analysis was performed using a Thermo Q Exactive PLUS equipped with a HESI source. The spray voltage was -2.7 kV in negative mode and 3.5 kV in positive mode. Sheath gas, auxiliary gas, and sweep gas flow rates were set at 40, 10, and 2 (arbitrary units), respectively. The capillary temperature was 300 °C, and the auxiliary gas heater was set to 360 °C. The S-lens RF level was 45, with an m/z scan range of 100 to 1200 m/z for both positive and negative ionization modes. Metabolite data was processed using the MAVEN software package 5 with a mass accuracy window of 5 ppm. For the lipidomics data analyses, the normalized mean values of Diacylglycerides (DAG) and Triglycerides (TAG) for fly heads from wildtype and tty-/- mutants from 7 biological replicates each were used. Normalization was achieved by comparing the response signal of target lipids and their respective internal standards, i.e. 5:0-18:1(d7) DAG for diacylglycerides and 5:0-18:1(d7)-15:0 TAG for triglycerides. Individual species from the lipid family were reported only when they were detected above the limit of quantitation (signal/noise = 10:1).
C1P quantitation by enzyme-linked immunosorbent assay (ELISA)
C1P was measured using a colorimetric C1P ELISA kit (AMS.E4332hu; Amsbio), which consists of anti-C1P antibody pre-coated well plate, biotinylated anti-C1P antibody, and streptavidin-HRP. Lipids were solvent-extracted from the cell lysates and vacuum dried. The dried lipids were dissolved in the standard diluent provided by the ELISA kit. After completing the colorimetric reaction, optical density (OD; 450 nm) of each sample was measured using a microplate reader. Individual OD values were normalized to the mean values of respective control samples.
Triglyceride quantitation assay
Triglyceride (TAG) contents were measured using an enzyme-coupled colorimetric kit (10010303; Cayman). For astrocytes, cells were seeded in 6-well plate at around 1 million cells/well or 24-well plate at around 25k cells/well. After respective treatments, cells in each well were washed with PBS before being scraped into 100 µL (6-well) or 50 µL (24-well) of standard diluent assay buffer provided by the kit. For fly head lipid extracts, each extract (from 30 fly heads) was reconstituted in 60 µL of standard diluent assay buffer. After completing the colorimetric reaction, optical density (OD; 540 nm) of each sample was measured using a microplate reader. Individual OD values were normalized to the mean values of respective control samples.
Quantitation of protein and TAG contents in mouse cortical tissue
Two-month-old Aldh1l1-Cre/ERT2; Ttyh1fl/fl mice received intraperitoneal injections of either corn oil (vehicle) or tamoxifen-containing (75 mg/kg-mouse) corn oil everyday for 5 consecutive days. Three days after the last injection, brain cortices were dissected out from euthanized mice. Each cortical tissue sample was homogenized in 1 mL of standard diluent assay buffer (700732; Cayman) using a Dounce homogenizer. Protein concentration in the tissue lysate was measured using Bradford reagent (5000205; Bio-Rad). Lipids in the tissue lysate were extracted using MTBE solvent phase separation. Extracted lipids were vacuum-dried and reconstituted in standard diluent assay buffer (700732; Cayman). TAG contents were measured by the triglyceride quantitation assay.
Protein-lipid in silico docking
The protein structure of human TTYH1 (accession number 7P5J) was obtained from the Protein Data Bank (PDB). The lipid structures were collected from the LIPID MAPS Structure Database 6. The structure of CerP(d18:0/22:0) was manually constructed in ChemDraw software. The ligands were prepared using the LigPrep tool by protonating molecules at a pH of 7±1 using Epik tool, minimizing the energy with the OPLS3e force field, and generating the tautomeric conformers. CB-Dock2 was used to identify the coordinates of the TTYH1 surface cavity and the constituent amino acid residues by matching to the hydrophobic cavity reported in recent cryo-EM studies 7,8. Isomeric chain A (7P5J-A) was prepared using ProPrep tool of Schrödinger where high-confidence protein structures were generated through adding missed hydrogen atoms, filling the missed side-chain atoms, generating the het states at pH: 7±2, deleting the het & water molecules (within 5.0 Å), assigning Hydrogen bonds using PROPKA pH:7 and converging the heavy atoms to root mean square deviation (RMSD) at greater than 0.30 Å. The receptor bound inhibitor molecule was chosen to generate GRID for the ligand binding using the GRID-GLIDE module. The constituent residues of the cavity, including Phe 33, Pro 35, Tyr 40, Leu 44, Thr 115, Val 119, Leu 122, Leu 126, Val 202, Asn 205, Val 206, Val 209, Glu 210, Arg 213, Tyr 217, Ser 260, Leu 263, Glu 264, Ala 265, Ala 266, Thr 267, Gly 270, leu 271, Phe 274, Tyr 281, Val 282, Leu 285, Thr 286, Tyr 377, Ala 380, Leu 381, Leu 384, Ala 388 and Leu 392 were picked to define the grid centroid to the active site. To minimize the penalties, the van der Waals radii were scaled at 0.80 and the default partial cut-off charge was set at 0.15. Using the GLIDE module, each ligand was docked with its corresponding protein binding site. The Extra precision (XP) module and OPLS3e force field were used for GLIDE-docking. The Epik state penalties were added to XP GLIDE score function. The Glide score was used to align the compounds based on their specific interactions to the proteins, e.g. hydrogen bonding. The top scored docking poses were visualized with PyMol graphics system v3.0, Schrödinger. The lipid ligands were represented in stick model. The constituent residues of the cavity were displayed in Figure 2 with surface representations: hydrophobic interacting residues were shown in green; residues E210, R213 and Y217 were shown in orange; and the rest were shown in grey.
Data presentation and statistical analyses
Comparisons between two groups were performed using two-tailed Student’s t-test, Mann-Whitney test, or ANOVA. Excel (Microsoft) and Prism 8 (GraphPad) were used for statistical analyses. Statistical significance was defined as P < 0.05. P-values are shown as asterisks in the figure: *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Graphs were generated in Prism 8 (GraphPad). Boxplots show median, interquartile range (box), and data range (whiskers). Violin plots show median (solid line), interquartile range (dotted line), and probability density of data (smoothed shape). Bar charts show mean average (bar) and standard error mean (whiskers). Data points on bar charts represent values from individual cells, individual Western blots, individual lipid extracts, or biological replicates. Distinct biological samples were used for Western blot and lipid quantitation assays. The sample sizes for all experimental conditions shown in the figures are summarized in Table 1.
References for Materials and Methods:
- Tcw, J. et al. An Efficient Platform for Astrocyte Differentiation from Human Induced Pluripotent Stem Cells. Stem Cell Reports 9, 600-614, doi:10.1016/j.stemcr.2017.06.018 (2017).
- Leung, H. H. et al. Drosophila tweety facilitates autophagy to regulate mitochondrial homeostasis and bioenergetics in Glia. Glia 72, 433-451, doi:10.1002/glia.24484 (2024).
- Jang, M. H. et al. Secreted frizzled-related protein 3 regulates activity-dependent adult hippocampal neurogenesis. Cell Stem Cell 12, 215-223, doi:10.1016/j.stem.2012.11.021 (2013).
- Karagas, N. E. et al. Loss of Activity-Induced Mitochondrial ATP Production Underlies the Synaptic Defects in a Drosophila Model of ALS. J Neurosci 42, 8019-8037, doi:10.1523/JNEUROSCI.2456-21.2022 (2022).
- Melamud, E., Vastag, L. & Rabinowitz, J. D. Metabolomic analysis and visualization engine for LC-MS data. Anal Chem 82, 9818-9826, doi:10.1021/ac1021166 (2010).
- Conroy, M. J. et al. LIPID MAPS: update to databases and tools for the lipidomics community. Nucleic Acids Res 52, D1677-D1682, doi:10.1093/nar/gkad896 (2024).
- Li, B., Hoel, C. M. & Brohawn, S. G. Structures of tweety homolog proteins TTYH2 and TTYH3 reveal a Ca(2+)-dependent switch from intra- to intermembrane dimerization. Nat Commun 12, 6913, doi:10.1038/s41467-021-27283-8 (2021).
- Sukalskaia, A., Straub, M. S., Deneka, D., Sawicka, M. & Dutzler, R. Cryo-EM structures of the TTYH family reveal a novel architecture for lipid interactions. Nat Commun 12, 4893, doi:10.1038/s41467-021-25106-4 (2021).