Antibodies
Flag tag [PA1-984B, Invitrogen. 1:1000-2000 (WB)]; HA tag [12013819001, Roche 1:5000 (WB)]; Actin [A3854, Sigma-Aldrich, 1:10000 WB]; V5 tag [13202, Cell Signaling, 1:2000 (WB)], CD45 [70257, Cell Signaling, 1:100 (IHC, IF)], SelO [ERP11968, Abcam, 1:2000 WB]; HSP60 [SC-13115, Santa Cruz Biotechnology, 1:25 IF]; TOM20 [A19403, ABclonal, 1:100 IF]; HADHA [10758-1-AP, Protein tech, 1:1000 WB]; HRP-linked goat anti-rabbit [7074, Cell Signaling, 1:10000 (WB)]; HRP-linked goat anti-rabbit [GK500705, GeneTech (Shanghai), ready-to-use diluted solution (IHC)]; HRP-linked horse anti-mouse [7076, Cell Signaling, 1:10000 (WB)]; goat anti rabbit-Alexa Fluor 633 [A21071, Invitrogen, 1:500 (IF)]; goat anti-rabbit Alexa Fluor 488 [A11008, Invitrogen, 1:500 (IF)] Goat anti mouse-Alexa Fluor 488 [A11001, Invitrogen, 1:500 (IF)]
The AMPylation antibody (7C11) was a kind gift from Prof. Aymelt Itzen from Universitätsklinikum Hamburg-Eppendorf.
siRNA
All the siRNAs were purchased from Synbio Technologies.
The sequences of the siRNAs were:
siCont:
sense: UUCUCCGAACGUGUCACGUdTdT
antisense:ACGUGACACGUUCGGAGAAdTdT
siSelO:
sense: CGCUGUUCUUCAGCGGCAAdTdT
antisense: UUGCCGCUGAAGAACAGCGdTdT
siSelO-2 (targeting 3’-UTR):
sense: AGUUGCUGGUUUAUUUAGGAGCCUGdTdT
antisense: CAGGCUCCUAAAUAAACCAGCAACUdTdT
siHADHA:
sense: UGUGGCAGUUGUUCGAAUUAAdTdT
antisense: UUAAUUCGAACAACUGCCACAdTdT
siABHD10:
sense: CCCUGGCUAUCUUUCUUAUAUdTdT
antisense: AUAUAAGAAAGAUAGCCAGGGdTdT
siDAP3:
sense: CCCGAGGAAUUAGCACUUGUUdTdT
antisense: AACAAGUGCUAAUUCCUCGGGdTdT
siSLC25A32:
sense: GCAGCAACAUACCCAUAUCAAdTdT
antisense: UUGAUAUGGGUAUGUUGCUGCdTdT
siFOXRED1:
sense: GCAGUUCUCAUUGCCUGAGAAdTdT
antisense: UUCUCAGGCAAUGAGAACUGCdTdT
siMETTL15:
sense: AGCCAGGCAGAAGCCUUAUUAdTdT
antisense: UAAUAAGGCUUCUGCCUGGCUdTdT
siOSGEOL1:
sense: CCAAGUGACCUCUCAGCAAUUdTdT
antisense: AAUUGCUGAGAGGUCACUUGGdTdT
siLYRM2:
sense: UGAUGAUUACUCAAGGCAAUAdTdT
antisense: UAUUGCCUUGAGUAAUCAUCAdTdT
siSTARD7:
sense: CGGUUGGAAGAAAUGUCAAAUdTdT
antisense: AUUUGACAUUUCUUCCAACCGdTdT
Chemicals and Reagents
NAD+, AMP, ADP, and ATP were purchased from Sangon Biotech. NADH, NMN, dibutylammonium acetate (DBAA), and Adenosine 2’,5’-diphosphate were from MACKLIN. NMNH was purchased from Bidepharm. 3-Ketopalmitoyl-CoA was from Toronto Research Chemicals. Oleic acid, palmitic acid, and methyl tert-butyl ether (MTBE) were from HEOWNS. Stearoyl coenzyme A was bought from Shanghai Yuanye Bio-technology Co., Ltd. Isotope labeled NAD+ was from Cambridge Isotope Laboratories. Palmitoyl-CoA was from Sigma-Aldrich. Oil Red O was from Solarbio, and BODIPY 493/503 was from MCE. Opti-MEM and DMEM were from Gibco. 100X penicillin-streptomycin, 100X non-essential amino acids, and doxycycline were from Solarbio. Fetal bovine serum was from Gibco.
Cell Lines and Animals
HEK293T, Hepa1-6, HepG2, and HeLa were from ATCC.
The SelO global knockout mice (SelO-/-) and the SelO floxed (SelOfl/fl) mice were purchased from Cyagen. The SelO floxed (SelOfl/fl) mice were crossed with Alb-Cre transgenic mice to generate liver-specific SelO knockout mice. All animals were kept under specific pathogen free (SPF) environment with 12 h light/12 h dark cycle. All animal experiments were approved by the Ethics Committee of Tianjin Medical University.
Plasmids
The plasmid for expression of SUMO-SelO (human) in E. coli was a kind gift from Prof. Vincent S. Tagliabracci from UT Southwestern. The plasmids encoding mitochondrial NAD sensors (186791), mitochondrial localized ratiometric pH sensor pHluorin (163053), and cytosolic super ecliptic pH sensor pHluorin (32001) were purchased from Addgene. The cytosolic super ecliptic pH sensor pHluorin was used as a template for cloning mitochondrial localized super ecliptic pHluorin,
The plenti-GFP-Flag was purchased from OBio Tech and served as the backbone vector for generating all lentiviral plasmids tagged with Flag, as well as plenti-GFP-HA and plenti-GFP-V5, which were used as backbone vectors for generating all the lentiviral plasmids tagged with HA and V5 tag, respectively.
The plasmids below were constructed for this study as described in Table S1:
pet28a-His6-ydiU-Flag, pLKO-tet-shSelO, plenti-SelO-Flag (mouse), pet28a-fadA-V5, pet28a-fadB-HA, pet28a-yfcX-HA, plenti-HADHB-V5, plenti-HADHA-HA, plenti-ACAA2-Flag, plenti-HADH-Flag, plenti-ECHS1-Flag, plenti-SelO-HA, pet28a-GST-ydiU, pet28a-ACOX1, mito-mCherry-SEpHluorin, pet28a-SUMO-SelO (C667A), pet28a-SUMO-SelO (R253E).
All homemade plasmids were validated by sequencing at Genewiz. For overexpression of SelO protein, we mutated the codon for selenocysteine to cysteine.
In silico Screening of NAD+ Binding Proteins
We employed three molecular docking programs, Vina, Autodock 4, and Ledock, to screen potentially uncharacterized NAD+ binding proteins in mitochondria. The subcellular localization information for all proteins in the human proteome was retrieved from the UniProt Database. Additionally, we identified known NAD+, NADH, NADP+, or NADPH binding proteins based on catalytic activity, binding site, and cofactor information retrieved from the UniProt database. Reverse blind docking was used for the evaluation of the binding affinities of each protein to NAD+ with AlphaFold2 modeled structures. These structures were downloaded from the AlphaFold2 database, except for the selenium-containing proteins, which were generated in situ using the AlphaFold2 algorithm upon replacing selenocysteine with cysteine in the sequence. This is necessary because the AlphaFold2 algorithm does not recognize residue abbreviation other than the twenty common amino acids, and structures for selenium-containing proteins are absent from the online database. Prior to molecular docking, we trimmed the intrinsically disordered regions in the AlphaFold2 modeled structures and truncated the structures into individual domains using the Leiden graph-clustering method based on the predicted alignment error matrices generated along with each AlphaFold2 modeled structure. The individual domains lacking sufficient topological complexity (alpha helix + beta strand ≤ 3) were discarded. For evaluation of the binding affinities, the molecular docking programs autonomously determined the binding sites by searching against the whole protein surface. Default parameters were used for all three molecular docking programs, with the exception of Vina, where search exhaustiveness was increased from the default value of 8 to 12. The versions for the docking programs were as follows: Vina: 1.2; Autodock 4: 4.2; Ledock 1.0. We evaluated the binding affinities towards NAD+ for all mitochondrial proteins, excluding known NAD+/NADH or NADP+/NADPH binding proteins, as well as known intracellular or extracellular NAD+ binding proteins (not confined to mitochondrial proteins). The average scores for known NAD+ binding proteins were used as the cutoffs for the selection of potentially uncharacterized NAD+ binding proteins in mitochondria.
HPLC-UV Analysis of the Enzymatic Reaction Products
Recombinantly purified SelO or ydiU were incubated with NAD+ or NADH in a buffer consisting of 50 mM Hepes (pH=8.2), 150 mM NaCl, 1 mM MnCl2, and 1 mM TCEP at 30 oC, and then the proteins were removed prior to HPLC analysis. For the reaction involving NAD+, the enzyme was removed by acid precipitation, followed by centrifugation to eliminate the precipitation. Subsequently, the solution was neutralized with NaOH before undergoing HPLC analysis. For the reaction involving NADH, enzyme removal was achieved through heat inactivation followed by centrifugation, because NADH is unstable when exposed to strong acid or base. The resulting reaction products were separated using an Agilent 1100 system coupled with an Agilent 1200 diode array detector equipped with deuterium and tungsten lamps for measurement of the absorbance. An HC-C18 column (5 μm, 4.6 X 250 mm) was employed for the separation. The eluents were (A) 100% methanol and (B) an aqueous solution containing 40 mM KH2PO4 and 60 mM K2HPO4, pH=7.0. The separation and subsequent analysis with HPLC-UV were conducted at room temperature. Isocratic elution with 100% B was used for the reaction involving NAD+, and gradient elution was used for the reaction involving NADH (solvent gradient: 0-10 min: 100% B; 10-20 min 100%-80% B). The quantification of AMP and NMN was based on absorbance measurements at 260 nm, while the quantification of NMNH relied on absorbance measurements at 340 nm. Note that NMNH does not exhibit absorbance at 260 nm.
Recombinant Protein Purification
Bacterial expressing vectors harboring fadB or fadA were transformed into BL21(DE3) cells. Expression vectors carrying wildtype or mutant ydiU or SUMO-tagged SelO were transformed into BL21(DE3) ΔydiU cells. The ydiU gene was knocked out to prevent potential formation of chimeric heteromultimer between endogenous ydiU and the induced proteins. The bacteria were cultured in LB medium at 37 oC until reaching an OD600 of 0.5-0.9. Protein expression was induced by adding 0.5 mM isopropyl-β-D-thiogalactoside and incubating for 12-16 h at 16 oC. Following induction, the bacterial cells were harvested and sonicated in lysis buffer (50 mM NaH2PO4 (pH 7.0), 0.1% Triton X-100, 10 mM imidazole, 5 mM BME, and 1 mM PMSF). The lysate was then centrifuged at 10,000 g for 10 min. Nucleic acids in the supernatant were precipitated by gradually adding streptomycin sulfate (final concentration w/v 2%), followed by another centrifugation step. The resulting supernatant was loaded onto a cobalt column (TALON). After washing with five bed volume of wash buffer (50 mM NaH2PO4 (pH 7.0), 0.1% Triton X-100, 20 mM imidazole, 5 mM BME, and 800 mM NaCl), proteins were eluted from the column using elution buffer (50 mM NaH2PO4 (pH 7.0), 250 mM imidazole, 1 mM BME and 300 mM NaCl). The eluate was further concentrated using Amicon Ultra and buffer-exchanged using Sephadex G-25 into a storage buffer containing 50 mM Hepes, 100 mM NaCl, 10% glycerol, and 1 mM DTT.
Measurement of NAD+ and Related Nucleotides from in vitro Enzymatic Reactions or Mitochondria using Targeted UPLC-MS/MS
For in vitro enzymatic reactions, the reaction was terminated by the addition of 1 M HCl to adjust the pH to 1-2, followed by neutralization with 1 M NaOH solution. For measurement of NAD+ levels in mitochondria, mitochondria were extracted from mouse liver using the Tissue Mitochondria Isolation Kit from Beyotime (C3606). The mitochondria were then suspended in mitochondrial respiration medium MiR05 buffer and incubated for the indicated time, followed by metabolite extraction with 80% methanol and sonication. The reaction mixtures from in vitro reactions or mitochondrial extracts were filtered through a 0.22 μm pore centrifuge tube filter (Costar 8169) prior to analysis on an ultra-performance liquid chromatography system equipped with a BEH T3 column (1.7 μm, 2.1 X 100 mm). The eluents were (A) an aqueous solution containing 0.25 mM di-n-butylamine acetate (DBAA) and (B) acetonitrile containing 3 mM DBAA. The separations were conducted at room temperature using gradient elution (solvent gradient: 0-6.5 min, 0-35% B). Targeted profiling of NAD+ and related nucleotides was performed with a hybrid triple quadrupole linear ion trap mass spectrometer (5500 QTRAP, AB SCIEX) equipped with an electrospray ionization source. The mass-to-charge ratios (m/z) were determined in negative mode. The concentrations of the analytes were detected by multireaction monitoring monitoring.
Surface Plasmon Resonance
SPR data were acquired on a Biacore 8K device (GE Healthcare). Recombinantly purified ydiU or SelO proteins were covalently immobilized on a CM5 sensor chip series S. The mobile phase was 10 mM Hepes (pH=7.5), 150 mM NaCl, 0.05% Tween-20. Serial dilutions of NAD+ were injected into flow cells at concentrations ranging from 12.5-1600 μM, with a flow rate of 30 μL/min at 25 oC. Each binding cycle consisted of an association phase of 120 s and a dissociation phase of 160 s. The maximal response units at each concentration were fitted to a 1:1 binding equation to determine the dissociation constants (Kd).
Quantification of Cellular and Mitochondrial NAD Levels using Colorimetric Methods
Cellular NAD levels (sum of NAD+ and NADH) were quantified using the WST-8 colorimetric method with a commercial kit (Beyotime S0175). Cell lysates were prepared using the buffer provided by the kit, and the total NAD levels were assessed by the formation of formazan through catalysis with excess alcohol dehydrogenase. For determination of the mitochondrial NAD levels, mitochondria were first isolated from the cells using a mitochondrial isolation kit (Beyotime C3601). Absorbance readings at 450 nm were taken with the Tecan Spark microplate reader at a slit width of 3.5 nm. Background absorbance at 450 nm was subtracted using the absorbance of a reaction mixture set up in the absence of cell lysate. The protein concentrations of the lysates were determined using the BCA method for the normalization of the cellular NAD levels.
Western Blot
Cells were lysed in PBS containing 0.5% Triton X-100 and a protease inhibitor cocktail by repeated freeze-and-thaw and vortexing with glass beads. Protein samples were separated by SDS-PAGE and transferred to the PVDF membrane. After blocking with 10% fat-free milk, membranes were incubated with primary antibodies overnight at 4 oC with gentle shaking, followed by a one-hour incubation with horseradish peroxidase-conjugated secondary antibodies at room temperature. All the primary and secondary antibodies were diluted in 1% fat-free milk in 1XTBST except for those used for the detection of AMPylation. For detection of AMPylation, the primary and secondary antibodies were diluted in protein-free blocking buffer (Pierce, 37585) supplemented with MnCl2. The PVDF membrane was also blocked with the protein-free blocking buffer for the detection of AMPylation. Blots were developed by chemiluminescence with ECL substrates from NCM Biotech. Immunoblots were visualized using the Tanon 5200 chemiluminescence imaging system.
Stable Cell Line Generation
HEK293T cells were co-transfected with pLKO-shSelO, packaging plasmid psPAX2, envelope plasmid pMD2.G using TRANIT-LT1. The supernatant containing lentivirus particles was collected and filtered through a sterilized 0.8 μm filter to remove floating cells or cell debris. HeLa cells were then infected with the lentivirus in the presence of polybrene and selected with 2 μg/ml puromycin for at least three days. To increase homogeneity within cell lines, monoclonal cell lines were generated by isolating single colonies from culture plates seeded at very low concentrations. The tetracycline-inducible expression of shSelO was verified by Western Blot analysis.
Flag Pulldown Assay
For pulldown with transfected mammalian cells, cells were lysed in PBS containing 0.5% Triton X-100 and a protease inhibitor cocktail by repeated freeze-and-thaw cycles and vortexing with glass beads. For pulldown with transformed E. coli, the bacterial cells were disrupted by sonication. After disrupting the cell pellets (either mammalian cells or bacterial cells), the lysates were centrifuged at 10,000g for 10 min at 4 oC, and the supernatant was loaded onto the Flag M2 magnetic beads. After incubation, the beads were washed three times with PBS. The bound proteins were then eluted with PBS containing 0.15 mg/mL 3XFlag peptide. The eluted proteins were either subjected to Western Blot analysis or mass spectrometry analysis. The mass spectrometry analysis, including tryptic digestion, peptide desalting and separation, mass spectra collection, and protein identification through database search, was conducted by the proteomic facility of Tianjin Medical University. An Orbitrap Q-extractive Plus mass spectrometer was used for the mass spectrometry analysis.
Size Exclusion Chromatography
Size exclusion chromatography analysis was carried out on an Agilent 1100 system equipped with an Agilent 1200 diode array detector for absorbance measurement. The column used was Superdex 200 Increase 10/300 GL column. Elution volume was estimated based on UV absorbance at 280 nm and 260 nm. The mobile phase consisted of 50 mM Hepes (pH=7.5) and 150 mM NaCl. The flow rate was 0.5 ml/min. For each analysis, we used 80-100 μl of protein solution at a concentration of approximately 10 μM. The proteins separated by size exclusion chromatography were collected using an automatic sampler, with one fraction collected every one minute. The proteins in each fraction were precipitated by adding three volumes of acetone and then redissolved in 1XLaemmli buffer. The samples were resolved by SDS-PAGE and stained with silver stain for analysis.
Generation of ydiU Knockout E.coli Cells
The ydiU gene was knocked out in BL21(DE3) E. coli using the λ RED recombinase system. First, BL21(DE3) cells were transformed with the helper plasmid pKD46, which encodes the RED recombinase. Subsequently, the transformed E. coli cells were again transformed with a PCR product containing an FRT-flanked kanamycin-resistant gene, with extensions on each end homologous to the sequence immediately before the start codon and after the stop codon of the ydiU gene. This PCR product was amplified from the pKD4 plasmid using the following primers:
ydiU_knockout_fwd:GACGAGAGTAACCGTCTACACTATCAAACAGGAGGATCTGTGTAGGCTGGAGCTGCTTC
ydiU_knockout_rev:AAAACTCAGGCTGGCAAGCTGCTGTTGACCAAGTAGCCTCATATGAATATCCTCCTTAG
Then, the cells were selected with kanamycin for recombination of the resistant gene at the locus of ydiU. The resistant gene was removed by transformation with the pCP20 plasmid, which encodes the FLP recombinase. Both the pKD46 and pCP20 plasmids contain temperature-sensitive replicons, enabling easy removal of the plasmids by raising the growing temperature.
Metabolomic Analysis of Cultured Cells
HeLa cells expressing doxycycline-inducible SelO targeting shRNA were treated with either doxycycline or vehicle for 48 h prior to cell harvesting and counting. Approximately 5X106 cells were used for each replicate. The cells were washed with PBS, and metabolites in the cells were extracted using a mixture of methanol, acetone, and water (2:2:1, v/v/v). The mixture was centrifuged at 14,000 g for 20 min, and the supernatant was then vacuum-dried. For LC-MS analysis, the samples were re-dissolved in a mixture of acetonitrile and water (1:1, v/v). The analysis was performed using a UPLC (1290 Infinity LC) equipped with a BEH Amide column (1.7 μm, 2.1 X100 mm) and coupled to a quadrupole time-of-flight mass spectrometer at Shanghai Applied Protein Technology Co., Ltd. The eluents were (A) an aqueous solution containing 25 mM NH4OAc and 25 mM NH3∙H2O, and (B) acetonitrile. The separations were conducted at room temperature using gradient elution (solvent gradient: 0-0.5 min, 95% B; 0.5-7 min, 96-65% B;7-8 min 65-40% B; 8-9 min 40 % B). Both ESI positive and negative modes were scanned. The data was processed with the XCMS software.
Drug Screening of SelO inhibitors
Drug screening was performed using the Explorer G3 integrated workstation, which includes a Janus G3 workstation equipped with a pin tool for dispensing compounds. Enzyme activity was measured using an Envision 2105 microplate reader, integrated into the system. A kinase inhibitor library from TargetMol was employed for the screening. We quantified the SelO activity based on the amount of AMP produced during NAD+ hydrolysis in a given time period. AMP is an inhibitor of firefly luciferase, which catalyzes a chemiluminescent reaction using luciferin and ATP as substrates. The amount of AMP produced was determined by assessing the inhibition of the firefly luciferase-mediated chemiluminescent reaction. Recombinantly purified SelO protein was first added to each well, followed by the addition of test compounds using the pin tool. After a 5-minute incubation, NAD+ was added, and the reaction was allowed to proceed for 2 hours. Subsequently, luciferase and its substrates were introduced into each well. Chemiluminescence was measured using the Envision 2105 microplate reader. Relative inhibitory activity was calculated by comparing the results from control wells (without inhibitors) and wells without SelO protein on the same microplate.
RNA-seq
The liver tissues were ground to a fine powder in liquid nitrogen, which was then transferred into TRIzol. Following this, the tissue samples underwent centrifugation at 12,000 g for 5 min at 4 oC, and then a mixture of chloroform and isoamyl alcohol (24:1) was added to the resulting supernatant. After another centrifugation at 12,000g for 8 min, the aqueous layer was recovered. Total RNA was precipitated from the aqueous layer and subsequently washed with 75% cold ethanol twice. Library construction and RNA-sequencing were carried out using the BGISEQ-500 platform by BGI-SHENZHEN Co.
Measurement of HADHA/HADHB Activity in vitro
For the forward reaction, HEK293T cells were co-transfected with both HADHA-Flag and HADHB-V5. The MTP complex was then immunopurified by pulldown with Flag M2 magnetic resin and elution with 3XFlag peptide. Palmitoyl-CoA (60 μM) was used as the starting material for the assay and was first converted into trans-hexadec-2-enoyl-CoA with excess recombinantly purified ACOX1 protein in O2 saturated buffer in situ. This is necessary because trans-hexadec-2-enoyl-CoA is not commercially available. After heat inactivation of the ACOX1 protein, and the reaction mixture was centrifuged at 10,000g for 10 min. The supernatant containing trans-hexadec-2-enoyl-CoA was directly used for the assay with HADHA and HADHB. The in vitro reaction was carried out with 250 μM NAD+ in the presence or absence of 1 μM recombinantly purified SelO, along with the purified HADHA and HADHB, at 30 oC for 2 h. The reaction was terminated by heat inactivation.
For the reverse reaction, HEK293T cells were transfected with HADHA-Flag, and the HADHA protein was immunopurified by pulldown with Flag M2 magnetic resin and elution with 3XFlag peptide, 3-ketopalmitoyl CoA (60 μM) was used as the starting material. The in vitro reaction was carried out with 250 μM NADH in the presence of absence of 1 μM recombinantly purified SelO, along with the purified HADHA, at 30 oC for 3 h. The reaction was terminated by heat inactivation.
After halting the reactions, stearoyl-CoA (30 μM) was supplemented into the reaction mixture as an internal control to account for the loss of fatty acyl-CoA molecules in the subsequent steps. The samples were vacuum-dried and then re-dissolved into a mixture of methanol and H2O (1:1). The re-dissolved samples were filtered through a 0.22 μm pore centrifuge tube filter (Costar 8169) prior to analysis on an ultra-performance liquid chromatography system equipped with a BEH C18 column (1.7 μm, 2.1 X 100 mm) The eluents were (A) an aqueous solution containing 5 mM NH4OAc and 2.5 mM DBAA and (B) 95% acetonitrile and 5% (A). The separations were conducted at room temperature using gradient elution (solvent gradient: 0-1 min, 2-50% B; 1-8.5 min, 50-98% B; 8.5-13.5 min 98% B). Non-targeted profiling of fatty acyl-CoA was performed with Thermo Orbitrap Exploris 480 equipped with an electrospray ionization source. The mass-to-charge ratios (m/z) were determined in positive mode. Thermo Xcalibur Qual Browser was used for data analysis. The formation of 3-ketopalmitoyl-CoA was used for quantifying the enzymatic activity in the forward reaction, and the formation of 3-hydroxylpalmitoyl-CoA was used for quantifying the enzymatic activity in the reverse reaction.
Measurement of the Hydrolysis of Isotope Labeled NAD+ with Isolated Mitochondria
Mitochondria were isolated from liver tissues obtained from wildtype mice or mice with liver-specific knockout of SelO using the Tissue Mitochondria Isolation Kit from Beyotime (C3606). Subsequently, the isolated mitochondria were resuspended in an oxygen saturated solution containing 20 mM NH4OAc (pH=7.45), 30 mM lactobionic acid, 20 mM taurine, 0.5 mM EGTA, 1 mM MgCl2, 1g/L BSA, and 1% Tween-20. Isotope labeled NAD+ (C13 labeling on the ribose linked to nicotinamide) was supplemented into the mixture and incubated at 30 oC for 1.5 h. Following incubation, metabolites were extracted with a mixture of methanol and acetone (1:1) using a Bioruptor pico sonication device. The solution was then centrifuged at 10,000 g for 10 min, and the supernatant was recovered and vacuumed dried. Subsequently, the samples were redissolved in ddH2O and filtered through a 0.22 μm pore centrifuge tube filter (Costar 8169) prior to analysis on an ultra-performance liquid chromatography system equipped with BEH C18 column (1.7 μm, 2.1 X 100 mm). The eluents were (A) an aqueous solution containing 10 mM NH4OAc (pH=9) (B) acetonitrile. The separations were conducted at room temperature using gradient elution (solvent gradient: 0-2 min, 3% B; 2-4 min, 3-90% B; 4-5 min 90% B). The hydrolysis of isotope labeled NAD+ was quantified by the formation of isotope labeled NMNH in the extract. The mass-to-charge ratios (m/z) were determined in positive mode.
Measurement of Blood Biochemical Parameters in Mice
For the measurement of HDL-C, LDL-C, ALT, AST, TC, and TG, blood was withdrawn from mice via cardiac puncture. The mice were anesthetized with isoflurane prior to blood collection. The blood samples were analyzed using an automatic biochemistry and immunoassay analyzer (Mindary SAL9000). The levels of ALT, AST, TC, and TG were determined using colorimetric enzymatic assays that employed lactate dehydrogenase, malate dehydrogenase, cholesterol oxidase, and triglyceride lipase, respectively. The HDL-C and LDL-C levels were determined using direct measurement methods. Ready-to-use reagent kits provided by the analyzer’s manufacturer were used. For measurement of blood glucose, tail blood was collected, and a Sinocare blood glucose meter was used along with the manufacturer-provided blood glucose test strips.
Measurement of Mouse Liver NEFA and TG Levels
Mouse tissues were mechanically homogenized using a bead mill homogenizer in either normal saline (for NEFA) or a 1:1 mixture of heptane and isopropanol (for TG). The resulting mixture was centrifuged at 10,000g for 10 min. The supernatant was collected for the determination of NEFA and TG levels. NEFA levels were assessed using a commercial kit from Nanjing Jiancheng Bioengineering Institute (A042-2-1). The levels were determined using an enzyme-based colorimetric assay employing acetyl-coA synthetase, acetyl-coA oxidase, and peroxidase. TG levels were assessed using a commercial kit from Solarbio (BC0620). The levels were determined using a chemical reaction-based colorimetric assay involving saponification of TG and oxidation of glycerol by periodic acid.
Oil Red Staining for Analysis of Lipid Content
Oil Red Staining was used to assess the levels of neutral lipids accumulated in cultured cells or liver tissues. Cultured cells were grown directly on microscopic slide before fixation with paraformaldehyde. Liver tissues were prefixed with 4% paraformaldehyde and then incubated in 20% and 30% sucrose sequentially for dehydration. The dehydrated tissues were cryosectioned and air-dried on precoated slides. The tissue slide or cell culture was fixed in 4% paraformaldehyde for 20 min at room temperature, followed by a brief wash with water. Subsequently, the slides were incubated with 60% isopropanol for 5 min before staining with Oil Red O working solution at room temperature for 20 min. After Oil Red staining, the slides were washed with water and stained with Hematoxylin. Images of the Oil Red Staining were captured using a Nikon Eclipse Ti microscope.
qPCR
For RNA extraction from liver tissues, the tissues were ground into fine powder in liquid nitrogen, and TRIzol reagent was added to the powder. After centrifugation for 10 min at 10,000g, the supernatant was used for total RNA extraction. For RNA extraction from cultured cells, the media was aspirated, and TRIzol reagent was directly added to the attached single-layer cells. Total RNA was isolated from the TRIzol solution following the manufacturer’s instructions. With the total RNA as template, cDNA was synthesized using a mixture of reverse transcriptase, oligo dT primer, and random primers (Transgene, AT301-03). Sybr Green master mix was used for PCR reactions as per the manufacturer’s instructions. qPCR assays were conducted using a BioRad CFX96 Real-Time PCR detection system. The sequence information for primer pairs used in qPCR is described in Table S2.
Measurement of fatty acyl-CoA in Mitochondria using Targeted UPLC-MS/MS
For measurement of fatty acyl-CoA levels in mitochondria, mitochondria were extracted from mouse liver using the Tissue Mitochondria Isolation Kit from Beyotime (C3606). Isolated mitochondria from approximately 600 mg liver tissues were suspended in 400 μl of 75% methanol. Metabolites were extracted by sonication, followed by centrifugation at 1,000 g for 10 min. The resulting supernatant was mixed with 1 ml of MTBE and incubated at room temperature with vortexing. Subsequently, around 250 μL of H2O was supplemented to the mixture and allowed to incubate for another 10 min. The mixture was then centrifuged at 12,000 g for 10 min, and the upper organic layer was collected and vacuum-dried for measurement of fatty acyl-CoA. The dried metabolites were redissolved in 80% methanol and filtered through a 0.22 μm pore centrifuge tube filter (Costar 8169) prior to analysis on an ultra-performance liquid chromatography system equipped with a BEH C18 column (1.7 μm, 2.1 X 100 mm). The eluents were (A) acetonitrile and (B) H2O. The separations were conducted at room temperature using gradient elution (solvent gradient: 0-2 min, 40% A; 2-7.5 min, 40-100 % A; 7.5-8 min 100% A). Targeted profiling of NAD+ and related nucleotides was performed with a hybrid triple quadrupole linear ion trap mass spectrometer (5500 QTRAP, AB SCIEX) equipped with an electrospray ionization source. The mass-to-charge ratios (m/z) were determined in positive mode.
Immunohistochemistry Staining of Liver Tissues
Human liver paraffin sections were prepared from freshly harvested tissues, which were fixed in 4% paraformaldehyde overnight and processed for paraffin embedding. The resulting paraffin blocks were sliced into 6 μm sections and mounted onto glass slides. The sections were deparaffinized with xylene, followed by rehydration through sequential immersion in alcohol solution of varying concentrations from 100% to 60%. The antigen unmasking was achieved with 10 mM sodium citrate (pH=6.0) buffer. The slides were then incubated with 3% H2O2 to quench endogenous peroxidase activity. Subsequently, the sections were blocked by 10% goat serum at room temperature for 1 h. After draining off the blocking buffer, the slides were incubated with the primary antibody solution containing 0.5% BSA at 4 oC overnight, followed by incubation with HRP conjugated secondary antibody solution at room temperature for 30 min. The antibody staining was visualized using DAB substrate solution, and the slides were then counterstained with Hematoxylin for 5 min. After dehydration with alcohol, the immunohistochemistry images were captured usinga Nikon Eclipse Ti microscope.
Fluorescence Microscopy
The fluorescence images for quantification of the mitochondrial NAD+ levels were obtained using live cells, the images for CD45 staining were obtained using liver paraffin sections, and the images for mitochondria were obtained using paraformaldehyde fixed cells. The liver parafiin sections were prepared following the same procedures as described in the immunohistochemistry staining section elaborated above, with the exception of using a fluorophore-conjugated secondary antibody instead of an HRP conjugated secondary one. Furthermore, an antifade mounting media (Beyotime, P0126) was applied to the microscope slides in the final step. For fixation of cultured cells, 2.5 % paraformaldehyde was used, followed by blocking and permeabilization with PBS buffer containing 3% BSA and 0.2 % Triton X-100. After permeabilization and blocking, cells were sequentially incubated with the primary and secondary antibody solutions diluted with PBS containing 1% BSA and 0.02% Triton-X 100. All fluorescent images were taken with a Zeiss LSM 800 confocal laser scanning microscope at room temperature using a 63X objective oil len.
Electron Microscopy Analysis of the Morphology of Mitochondria
Cells or tissues were fixed with 2.5% glutaraldehyde solution at 4 oC for 12 h. Subsequently, the samples were further fixed with 1% osmium tetroxide and dehydrated using a series of acetone solutions. Following dehydration, the samples were embedded in epoxy resin and then sectioned into 70-90 nm thin slides, which were then mounted on the specimen grid. The samples were then sequentially stained with uranyl acetate and lead citrate, followed by drying with filter paper. Electron microscopic images were captured using a Hitachi-800 Transmission Electron Microscope.
Statistical Analyses
Data were presented as mean ± SD or mean ± SEM as indicated in figure legends. Sample size N was indicated in figure legends. Statistical significance was set at p<0.05. n.s. stands for “not significant”. Significance between two groups was determined using Student’s t-test (unpaired, two-tailed, unequal variance).